Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Prev Med ; 180: 107888, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38325609

RESUMO

OBJECTIVE: Denmark went through various COVID-19 pandemic restrictions including periodic lockdowns from March 2020 to January 2022. All cancer screening programs were kept operational, yet access to clinicians for cervical screening was at times limited. We assessed the impact of the pandemic on cervical cancer screening activity in the Capital Region of Denmark. METHODS: Cervical screening activity was defined as regular screening by invitation, opportunistic screening, and screening participation by HPV self-sampling. Activity was monitored during and post-pandemic and compared relatively to a 3-year pre-pandemic reference. RESULTS AND CONCLUSIONS: The activity of cervical cancer screening was initially affected by the pandemic lockdowns, but increased activity during summer 2020 partly compensated this effect. Regular screening activity decreased 8.4% in 2020 and returned to pre-pandemic levels in 2021. During 2022 restrictions were removed and the decrease in activity was recorded to be 2.3%. Opportunistic screening activity was reduced by 14.3% in 2020 and 12.6% in 2021. A continued post-pandemic opportunistic screening activity reduction of 18.5% was observed, possibly related to changed patterns of primary health care use introduced during the pandemic. Screening by HPV self-sampling increased from 17.1% in the pre-pandemic period to 21.2% during the pandemic. Significantly more acceptance was recorded amongst older women (p < 0.0001). This increase mirrors the decrease in total clinician collected sample activity during the pandemic, where an increased reduction by age was observed. Post-pandemic HPV self-sampling participation decreased to 12.8%, possible reflecting a temporarily changed composition and motivation in the group of women invited for self-sampling.


Assuntos
COVID-19 , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Idoso , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/epidemiologia , Detecção Precoce de Câncer/métodos , Pandemias/prevenção & controle , Esfregaço Vaginal , Infecções por Papillomavirus/diagnóstico , Papillomaviridae , Autocuidado/métodos , COVID-19/diagnóstico , Controle de Doenças Transmissíveis , Manejo de Espécimes/métodos , Programas de Rastreamento/métodos , Dinamarca/epidemiologia
2.
Sci Rep ; 11(1): 21839, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750429

RESUMO

The intimate association between obesity and type II diabetes urges for a deeper understanding of adipocyte function. We and others have previously delineated a role for the tumor suppressor p53 in adipocyte biology. Here, we show that mice haploinsufficient for MDM2, a key regulator of p53, in their adipose stores suffer from overt obesity, glucose intolerance, and hepatic steatosis. These mice had decreased levels of circulating palmitoleic acid [non-esterified fatty acid (NEFA) 16:1] concomitant with impaired visceral adipose tissue expression of Scd1 and Ffar4. A similar decrease in Scd and Ffar4 expression was found in in vitro differentiated adipocytes with perturbed MDM2 expression. Lowered MDM2 levels led to nuclear exclusion of the transcriptional cofactors, MORC2 and LIPIN1, and thereby possibly hampered adipocyte function by antagonizing LIPIN1-mediated PPARγ coactivation. Collectively, these data argue for a hitherto unknown interplay between MDM2 and MORC2/LIPIN1 involved in balancing adipocyte function.


Assuntos
Tecido Adiposo Branco/metabolismo , Resistência à Insulina/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos Monoinsaturados/sangue , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Feminino , Redes Reguladoras de Genes , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Haploinsuficiência/genética , Haploinsuficiência/fisiologia , Resistência à Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , PPAR gama/metabolismo , Fosfatidato Fosfatase , Proteínas Proto-Oncogênicas c-mdm2/deficiência , Proteínas Proto-Oncogênicas c-mdm2/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
3.
Am J Physiol Endocrinol Metab ; 310(2): E116-28, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26578713

RESUMO

The tumor suppressor p53 (TRP53 in mice) is known for its involvement in carcinogenesis, but work during recent years has underscored the importance of p53 in the regulation of whole body metabolism. A general notion is that p53 is necessary for efficient oxidative metabolism. The importance of UCP1-dependent uncoupled respiration and increased oxidation of glucose and fatty acids in brown or brown-like adipocytes, termed brite or beige, in relation to energy balance and homeostasis has been highlighted recently. UCP1-dependent uncoupled respiration in classic interscapular brown adipose tissue is central to cold-induced thermogenesis, whereas brite/beige adipocytes are of special importance in relation to diet-induced thermogenesis, where the importance of UCP1 is only clearly manifested in mice kept at thermoneutrality. We challenged wild-type and TRP53-deficient mice by high-fat feeding under thermoneutral conditions. Interestingly, mice lacking TRP53 gained less weight compared with their wild-type counterparts. This was related to an increased expression of Ucp1 and other PPARGC1a and PPARGC1b target genes but not Ppargc1a or Ppargc1b in inguinal white adipose tissue of mice lacking TRP53. We show that TRP53, independently of its ability to bind DNA, inhibits the activity of PPARGC1a and PPARGC1b. Collectively, our data show that TRP53 has the ability to regulate the thermogenic capacity of adipocytes through modulation of PPARGC1 activity.


Assuntos
Tecido Adiposo Marrom/metabolismo , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Termogênese/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica , Feminino , Regulação da Expressão Gênica , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteína Desacopladora 1 , Aumento de Peso/fisiologia
4.
J Ovarian Res ; 7: 110, 2014 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-25432544

RESUMO

BACKGROUND: Corona radiata cells (CRCs) refer to the fraction of cumulus cells just adjacent to the oocyte. The CRCs are closely connected to the oocyte throughout maturation and their gene expression profiles might reflect oocyte quality. Polycystic ovary syndrome (PCOS) is a common cause of infertility. It is controversial whether PCOS associate with diminished oocyte quality. The purpose of this study was to compare individual human CRC samples between PCOS patients and controls. METHODS: All patients were stimulated by the long gonadotropin-releasing hormone (GnRH) agonist protocol. The CRC samples originated from individual oocytes developing into embryos selected for transfer. CRCs were isolated in a two-step denudation procedure, separating outer cumulus cells from the inner CRCs. Extracted RNA was amplified and transcriptome profiling was performed with Human Agilent® arrays. RESULTS: The transcriptomes of CRCs showed no individual genes with significant differential expression between PCOS and controls, but gene set enrichment analysis identified several cell cycle- and DNA replication pathways overexpressed in PCOS CRCs (FDR < 0.05). Five of the genes contributing to the up-regulated cell cycle pathways in the PCOS CRCs were selected for qRT-PCR validation in ten PCOS and ten control CRC samples. qRT-PCR confirmed significant up-regulation in PCOS CRCs of cell cycle progression genes HIST1H4C (FC = 2.7), UBE2C (FC = 2.6) and cell cycle related transcription factor E2F4 (FC = 2.5). CONCLUSION: The overexpression of cell cycle-related genes and cell cycle pathways in PCOS CRCs could indicate a disturbed or delayed final maturation and differentiation of the CRCs in response to the human chorionic gonadotropin (hCG) surge. However, this had no effect on the in vitro development of the corresponding embryos. Future studies are needed to clarify whether the up-regulated cell cycle pathways in PCOS CRCs have any clinical implications.


Assuntos
Blastocisto/metabolismo , Síndrome do Ovário Policístico/metabolismo , Transcriptoma , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Infertilidade Feminina/metabolismo , Infertilidade Feminina/terapia , Análise de Sequência com Séries de Oligonucleotídeos , Oócitos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Injeções de Esperma Intracitoplásmicas , Adulto Jovem
5.
Endocr Relat Cancer ; 19(3): 365-79, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22420006

RESUMO

Testicular germ cell tumours, seminoma (SE) and non-seminoma (NS), of young adult men develop from a precursor cell, carcinoma in situ (CIS), which resembles foetal gonocytes and retains embryonic pluripotency. We used microarrays to analyse microRNA (miRNA) expression in 12 human testis samples with CIS cells and compared it with miRNA expression profiles of normal adult testis, testis with Sertoli-cell-only that lacks germ cells, testis tumours (SE and embryonal carcinoma (EC), an undifferentiated component of NS) and foetal male and female gonads. Principal components analysis revealed distinct miRNA expression profiles characteristic for each of the different tissue types. We identified several miRNAs that were unique to testis with CIS cells, foetal gonads and testis tumours. These included miRNAs from the hsa-miR-371-373 and -302-367 clusters that have previously been reported in germ cell tumours and three miRNAs (hsa-miR-96, -141 and -200c) that were also expressed in human epididymis. We found several miRNAs that were upregulated in testis tumours: hsa-miR-9, -105 and -182-183-96 clusters were highly expressed in SE, while the hsa-miR-515-526 cluster was high in EC. We conclude that the miRNA expression profile changes during testis development and that the miRNA profile of adult testis with CIS cells shares characteristic similarities with the expression in foetal gonocytes.


Assuntos
Carcinoma in Situ/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Testiculares/genética , Testículo/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Ovário/metabolismo , RNA Mensageiro/genética
6.
Methods Mol Biol ; 755: 307-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21761315

RESUMO

Laser microdissection permits isolation of specific cell types from tissue sections or cell cultures. This may be beneficial when investigating the role of specific cells in a complex tissue or organ. In tissues with easily distinguishable morphology, a simple hematoxylin staining is sufficient, but in most cases a more specific staining is required to identify which cells to microdissect. We have established two staining protocols for frozen sections (1) Oil red O, which stains lipid droplet in fat cells and steroid-producing cells and (2) NBT BCIP, which stains cells expressing an alkaline phosphatase enzyme, such as fetal germ cells, testicular carcinoma in situ cells, and putatively also other early stem cell populations. We have applied these protocols for microdissection of rat Leydig cells, fetal human and zebrafish germ cells, and human testicular germ cell tumors, but the staining protocols could also be used in other species and for other cell types containing lipid droplets or expressing alkaline phosphatase. Both protocols ensure a morphology that enables microdissection of single cells with RNA quality sufficient for subsequent gene expression analysis. However, RNA yields after microdissection and purification are small, and therefore, two rounds of linear amplification are recommended prior to gene expression analysis.


Assuntos
Perfilação da Expressão Gênica/métodos , Lasers , Microdissecção/métodos , Testículo/metabolismo , Animais , Compostos Azo/química , Corantes/química , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA/genética , RNA/isolamento & purificação , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Coloração e Rotulagem/métodos , Fixação de Tecidos/métodos
7.
PLoS One ; 6(6): e21647, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738749

RESUMO

BACKGROUND: Polyunsaturated n-3 fatty acids (n-3 PUFAs) are reported to protect against high fat diet-induced obesity and inflammation in adipose tissue. Here we aimed to investigate if the amount of sucrose in the background diet influences the ability of n-3 PUFAs to protect against diet-induced obesity, adipose tissue inflammation and glucose intolerance. METHODOLOGY/PRINCIPAL FINDINGS: We fed C57BL/6J mice a protein- (casein) or sucrose-based high fat diet supplemented with fish oil or corn oil for 9 weeks. Irrespective of the fatty acid source, mice fed diets rich in sucrose became obese whereas mice fed high protein diets remained lean. Inclusion of sucrose in the diet also counteracted the well-known anti-inflammatory effect of fish oil in adipose tissue, but did not impair the ability of fish oil to prevent accumulation of fat in the liver. Calculation of HOMA-IR indicated that mice fed high levels of proteins remained insulin sensitive, whereas insulin sensitivity was reduced in the obese mice fed sucrose irrespectively of the fat source. We show that a high fat diet decreased glucose tolerance in the mice independently of both obesity and dietary levels of n-3 PUFAs and sucrose. Of note, increasing the protein∶sucrose ratio in high fat diets decreased energy efficiency irrespective of fat source. This was accompanied by increased expression of Ppargc1a (peroxisome proliferator-activated receptor, gamma, coactivator 1 alpha) and increased gluconeogenesis in the fed state. CONCLUSIONS/SIGNIFICANCE: The background diet influence the ability of n-3 PUFAs to protect against development of obesity, glucose intolerance and adipose tissue inflammation. High levels of dietary sucrose counteract the anti-inflammatory effect of fish oil in adipose tissue and increases obesity development in mice.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Óleos de Peixe/uso terapêutico , Obesidade/tratamento farmacológico , Sacarose/efeitos adversos , Tecido Adiposo/metabolismo , Animais , Calorimetria Indireta , Dieta Hiperlipídica/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/induzido quimicamente , Reação em Cadeia da Polimerase
8.
Cancer Res ; 69(12): 5241-50, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19491264

RESUMO

Testicular germ cell cancers in young adult men derive from a precursor lesion called carcinoma in situ (CIS) of the testis. CIS cells were suggested to arise from primordial germ cells or gonocytes. However, direct studies on purified samples of CIS cells are lacking. To overcome this problem, we performed laser microdissection of CIS cells. Highly enriched cell populations were obtained and subjected to gene expression analysis. The expression profile of CIS cells was compared with microdissected gonocytes, oogonia, and cultured embryonic stem cells with and without genomic aberrations. Three samples of each tissue type were used for the analyses. Unique expression patterns for these developmentally very related cell types revealed that CIS cells were very similar to gonocytes because only five genes distinguished these two cell types. We did not find indications that CIS was derived from a meiotic cell, and the similarity to embryonic stem cells was modest compared with gonocytes. Thus, we provide new evidence that the molecular phenotype of CIS cells is similar to that of gonocytes. Our data are in line with the idea that CIS cells may be gonocytes that survived in the postnatal testis. We speculate that disturbed development of somatic cells in the fetal testis may play a role in allowing undifferentiated cells to survive in the postnatal testes. The further development of CIS into invasive germ cell tumors may depend on signals from their postpubertal niche of somatic cells, including hormones and growth factors from Leydig and Sertoli cells.


Assuntos
Perfilação da Expressão Gênica , Neoplasias Testiculares/genética , Sequência de Bases , Primers do DNA , Humanos , Imunoquímica , Hibridização In Situ , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Testiculares/patologia
9.
PLoS One ; 4(5): e5536, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19436754

RESUMO

Microarray and RT-PCR based methods are important tools for analysis of gene expression; however, in tissues containing many different cells types, such as the testis, characterization of gene expression in specific cell types can be severely hampered by noise from other cells. The laser microdissection technology allows for enrichment of specific cell types. However, when the cells are not morphologically distinguishable, it is necessary to use a specific staining method for the target cells. In this study we have tested different fixatives, storage conditions for frozen sections and staining protocols, and present two staining protocols for frozen sections, one for fast and specific staining of fetal germ cells, testicular carcinoma in situ cells, and other cells with embryonic stem cell-like properties that express the alkaline phosphatase, and one for specific staining of lipid droplet-containing cells, which is useful for isolation of the androgen-producing Leydig cells. Both protocols retain a morphology that is compatible with laser microdissection and yield RNA of a quality suitable for PCR and microarray analysis.


Assuntos
Carcinoma in Situ/patologia , Lasers , Microdissecção/instrumentação , Coloração e Rotulagem/métodos , Neoplasias Testiculares/patologia , Testículo/citologia , Humanos , Masculino , Análise em Microsséries , Microdissecção/métodos , RNA/isolamento & purificação , RNA/metabolismo , RNA Neoplásico/isolamento & purificação , RNA Neoplásico/metabolismo , Testículo/metabolismo
10.
Mol Cell Endocrinol ; 288(1-2): 111-8, 2008 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-18420341

RESUMO

Carcinoma in situ (CIS) testis, known also as intratubular germ cell neoplasia, is the cancer stem cell from which the great majority of testicular germ cell derived tumours (TGCTs) of the testis arise. TGCTs can proliferate into morphologically homogeneous seminomas or can differentiate into virtually any type of tissue and form teratomas (non-seminomas). CIS cells display a close phenotypic similarity to fetal germ cells (primordial germ cells or gonocytes) suggesting an origin due to a developmental delay or arrest of differentiation of early germ cells. The pluripotency of these neoplasms has recently been explained by a close resemblance of their expression profile to that of embryonic inner cell mass cells studied in culture as embryonic stem cells, with high expression of transcription factors associated with pluripotency, such as NANOG and OCT3/4, as well as proteins found in several tissue specific stem cells, such as TFAP2C (AP-2gamma) or KIT. CIS and seminomas highly express a number of pre-meiotic germ cell specific genes, which are down-regulated during development to non-seminomas, while the expression of other embryonic markers, such as SOX2, is up-regulated. The mechanistic pathways and causative factors remain to be elucidated of both the initial transformation of fetal germ cells into CIS cells and the progression of CIS cells into an invasive tumour in the young adult. However, evidence supported by epidemiological studies indicate that disturbances in the hormonal microenvironment of the differentiating gonads may results in both the neoplasia and a host of other problems later in life, such as genital malformations, decreased spermatogenesis, and signs of hypogonadism.


Assuntos
Desenvolvimento Embrionário , Neoplasias Embrionárias de Células Germinativas/patologia , Animais , Biomarcadores/metabolismo , Carcinoma in Situ/patologia , Células-Tronco de Carcinoma Embrionário , Desenvolvimento Embrionário/efeitos dos fármacos , Disruptores Endócrinos/farmacologia , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia
11.
Int J Androl ; 31(2): 275-87, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18205797

RESUMO

Recent increases in male reproductive disorders have been linked to exposure to environmental factors leading to the testicular dysgenesis syndrome (TDS). Testicular cancer is the most severe condition in TDS and studies have shown a clear correlation between risk of testicular cancer and other components of TDS and that the geographical location of the mother during pregnancy can be a risk factor. This suggests that the dysgenesis has its origin in utero and that TDS is initiated by environmental factors, including possibly hormone-disrupting compounds that act on the mother and the developing foetus, but the genetic background may also play a role. The morphological similarity of carcinoma in situ (CIS) cells (the precursor of the majority of invasive testicular cancers) with primordial germ cells and gonocytes, and overlap in expression of protein markers suggests an origin of CIS from primordial germ cells or gonocytes. CIS cells and germ cell-derived cancers of the human type have so far not been described in any animal model of TDS, which could be caused by species differences in the development of the male gonad. Regardless of this, it is plausible that the dysgenesis, and hence the development of CIS cells, is a result of disturbed signalling between nurse cells and germ cells that allow embryonic germ cells to survive in the pre-pubertal and adult testis. The post-pubertal proliferation of CIS cells combined with aberrant signalling then leads to an accumulation of genetic changes in the CIS cells, which eventually results in the development of invasive testicular cancer in the adult.


Assuntos
Neoplasias Testiculares/etiologia , Testículo/patologia , Humanos , Masculino , Síndrome , Neoplasias Testiculares/patologia
12.
Biol Reprod ; 78(5): 852-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18199879

RESUMO

NANOG, POU5F1, and SOX2 are required by the inner cell mass of the blastocyst and act cooperatively to maintain pluripotency in both mouse and human embryonic stem cells. Inadequacy of any one of them causes loss of the undifferentiated state. Mouse primordial germ cells (PGCs), from which pluripotent embryonic germ cells (EGCs) are derived, also express POU5F1, NANOG, and SOX2. Thus, a similar expression profile has been predicted for human PGCs. Here we show by RT-PCR, immunoblotting, and immunohistochemistry that human PGCs express POU5F1 and NANOG but not SOX2, with no evidence of redundancy within the group B family of human SOX genes. Although lacking SOX2, proliferative human germ cells can still be identified in situ during early development and are capable of culture in vitro. Surprisingly, with the exception of FGF4, many stem cell-restricted SOX2 target genes remained detected within the human SOX2-negative germ cell lineage. These studies demonstrate an unexpected difference in gene expression between human and mouse. The human PGC is the first primary cell type described to express POU5F1 and NANOG but not SOX2. The data also provide a new reference point for studies attempting to turn human stem cells into gametes by normal developmental pathways for the treatment of infertility.


Assuntos
Linhagem da Célula/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/metabolismo , Proteínas HMGB/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Feminino , Células Germinativas/citologia , Proteínas HMGB/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Proteína Homeobox Nanog , Neoplasias Embrionárias de Células Germinativas/metabolismo , Neoplasias Embrionárias de Células Germinativas/patologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Fatores de Transcrição SOXB1 , Testículo/citologia , Testículo/embriologia , Testículo/metabolismo , Fatores de Transcrição/genética
13.
Best Pract Res Clin Endocrinol Metab ; 21(3): 462-78, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17875492

RESUMO

The testicular dysgenesis syndrome (TDS) hypothesis proposes that a proportion of the male reproductive disorders-cryptorchidism, hypospadias, infertility and testicular cancer-may be symptoms of one underlying developmental disease, TDS, which is most likely a result of disturbed gonadal development in the embryo. TDS may be caused by genetic factors, environmental/life-style factors, or a combination of both. Some rare disorders of sex development of genetic origin are among the best-known examples of severe TDS. Among the environmental and life-style factors that are suspected to influence the hormonal milieu of the developing gonad are the endocrine disrupters. A prenatal exposure to commonly used chemicals, e.g. phthalates, may result in a TDS-like phenotype in rats. Currently, this animal model is the best model for TDS. In humans the situation is much more complex, and TDS exists in a wide range of phenotypes: from the mildest and most common form, in which impaired spermatogenesis is the only symptom, to the most severe cases, in which the patient may develop testicular cancer. It is of great importance that clinicians in different specialties treating patients with TDS are aware of the association between the different symptoms.


Assuntos
Carcinógenos Ambientais/toxicidade , Disruptores Endócrinos/toxicidade , Disgenesia Gonadal/embriologia , Disgenesia Gonadal/genética , Neoplasias Testiculares , Animais , Carcinoma in Situ/induzido quimicamente , Carcinoma in Situ/embriologia , Carcinoma in Situ/epidemiologia , Carcinoma in Situ/genética , Feminino , Doenças dos Genitais Masculinos/embriologia , Doenças dos Genitais Masculinos/genética , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/induzido quimicamente , Neoplasias Embrionárias de Células Germinativas/embriologia , Neoplasias Embrionárias de Células Germinativas/epidemiologia , Neoplasias Embrionárias de Células Germinativas/genética , Ratos , Espermatogênese/fisiologia , Neoplasias Testiculares/induzido quimicamente , Neoplasias Testiculares/embriologia , Neoplasias Testiculares/epidemiologia , Neoplasias Testiculares/genética , Testículo/embriologia , Testículo/fisiopatologia , População Branca
14.
Virchows Arch ; 449(2): 200-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16736189

RESUMO

Testicular germ-cell tumours of young adults are derived from a pre-invasive intratubular lesion, carcinoma in situ (CIS). In a recent genome-wide gene expression screening using cDNA microarrays, we found PDPN over-expressed in CIS compared to normal adult testis. PDPN encodes podoplanin (Aggrus, human gp36, T1A-2), a transmembrane glycoprotein expressed in lymphatic endothelium and various solid tumours. To examine a potential role for PDPN in testicular neoplasms and during testicular development, we investigated its expression pattern during the development of human testis and in a series of testicular CIS, gonadoblastoma and overt germ-cell tumours. We established by RT-PCR and by immunohistochemistry with a gp36 antibody that PDPN mRNA and the protein product were expressed in testes with germ-cell neoplasms but not in the normal adult testis. We also found gp36 expression in early foetal gonocytes and immature Sertoli cells, similar to the expression pattern of M2A antigen, a previously identified marker for CIS and seminoma. This reinforced our previous proposal that M2A (D2-40) antigen was identical to gp36 (podoplanin, Aggrus, T1A-2). Our findings also suggest that podoplanin has a function in developing testis, most likely at the level of cell-cell interactions among pre-meiotic germ cells and immature Sertoli cells.


Assuntos
Antígenos de Neoplasias/análise , Carcinoma in Situ/química , Glicoproteínas de Membrana/análise , Neoplasias Embrionárias de Células Germinativas/química , Neoplasias Testiculares/química , Testículo/química , Adolescente , Adulto , Criança , Pré-Escolar , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Testículo/embriologia
15.
Int J Androl ; 29(1): 211-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16466542

RESUMO

Since the discovery of testicular carcinoma in situ (CIS) -- the precursor cell for the vast majority of germ cell tumours -- it has been proposed that CIS cells could be derived from transformed primordial germ cells or gonocytes. Here, we review recent discoveries not only substantiating that initial hypothesis but also indicating that CIS cells have a striking phenotypic similarity to embryonic stem cells (ESC). Many cancers have been proposed to originate from tissue-specific stem cells [so-called 'cancer stem cells' (CSC)] and we argue that CIS may be a very good example of a CSC, but with exceptional features due to the retention of embryonic pluripotency. In addition, considering the fact that pre-invasive CIS cells are transformed from early fetal cells, possibly due to environmentally induced alterations of the niche, we discuss potential risks linked to the uncontrolled therapeutic use of ESC.


Assuntos
Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco/patologia , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Germinoma/genética , Germinoma/metabolismo , Germinoma/patologia , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/metabolismo , Seminoma/genética , Seminoma/metabolismo , Seminoma/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
16.
Cell Tissue Res ; 322(1): 159-65, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15846514

RESUMO

Testicular cancer is the most common malignancy among men in the reproductive age and the incidence is increasing, probably caused by environmental factors. Most testicular cancers are testicular germ cell tumours and all originate from a carcinoma in situ (CIS) pattern. In this review, we focus on the pre-invasive CIS and its possible fetal origin by reviewing recent data originating from DNA microarrays and comparative genomic hybridisations. A comparison of gene expression and genomic aberrations reveal chromosomal "hot spots" with mutual clustering of gene expression and genomic amplification. Some of the genes found in the hot spots may be involved in creating the CIS phenotype. On the other hand, many genes that are highly expressed in CIS are not present in the hot-spot areas. The gene expression profile of CIS thus most likely reflects the combined result of genomic amplification and increased transcriptional activation and/or deficiency in the epigenetic silencing of specific loci. Amplification of chromosome 12p, appears to be a good genomic marker of the transition from the pre-malignant to malignant CIS cell; this is consistent with recent findings of propagation advantages in cultured undifferentiated embryonic stem cells after spontaneous amplification in similar regions. The gene expression profile of CIS cells has remarkable similarity to that of embryonic stem cells and supports our long-standing hypothesis of an early developmental origin of CIS and testicular germ cell cancer.


Assuntos
Carcinoma in Situ , Perfilação da Expressão Gênica , Genoma , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Carcinoma in Situ/etiologia , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Aberrações Cromossômicas , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/etiologia , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Estatística como Assunto , Neoplasias Testiculares/etiologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
17.
Clin Cancer Res ; 10(24): 8521-30, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15623634

RESUMO

PURPOSE: Transcription factor activator protein-2gamma (TFAP2C, AP-2gamma) was reported previously in extraembryonic ectoderm and breast carcinomas but not in the testis. In our recent gene expression study we detected AP-2gamma in carcinoma in situ testis (CIS, or intratubular germ cell neoplasia), precursor of testicular germ cell tumors. In this study we aimed to investigate the expression pattern of AP-2gamma and to shed light on this factor in germ cell differentiation and the pathogenesis of germ cell neoplasia. EXPERIMENTAL DESIGN: We analyzed expression pattern of AP-2gamma at the RNA and protein level in normal human tissues and a panel of tumors and tumor-derived cell lines. In the gonads, we established the ontogeny of expression of AP-2gamma in normal and dysgenetic samples. We also investigated the regulation of AP-2gamma by steroids and retinoic acid. RESULTS: We detected abundant AP-2gamma in testicular CIS and in testicular germ cell tumors of young adults and confirmed differential expression of AP-2gamma in somatic tumors. We found that AP-2gamma expression was regulated by retinoic acid in an embryonal carcinoma cell line (NT2). The investigation of ontogeny of AP-2gamma protein expression in fetal gonads revealed that it was confined to oogonia/gonocytes and was down-regulated with germ cell differentiation. In some prepubertal intersex cases, AP-2gamma was detected outside of the normal window of expression, probably marking neoplastic transformation of germ cells. CONCLUSIONS: AP-2gamma is developmentally regulated and associated with the undifferentiated phenotype in germ cells. This transcription factor may be involved in self-renewal and survival of immature germ cells and tissue-specific stem cells. AP-2gamma is a novel marker of testicular CIS and CIS-derived tumors.


Assuntos
Biomarcadores Tumorais , Carcinoma in Situ/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neoplasias Ovarianas/metabolismo , Neoplasias Testiculares/metabolismo , Fatores de Transcrição/metabolismo , Adolescente , Adulto , Carcinoma in Situ/genética , Diferenciação Celular , Criança , Pré-Escolar , Proteínas de Ligação a DNA/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Germinoma/etiologia , Germinoma/metabolismo , Disgenesia Gonadal/complicações , Disgenesia Gonadal/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Gravidez , Esteroides/farmacologia , Neoplasias Testiculares/genética , Fator de Transcrição AP-2 , Fatores de Transcrição/genética , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA