Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Biol Chem ; 299(4): 104570, 2023 04.
Article in English | MEDLINE | ID: mdl-36870679

ABSTRACT

Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies have uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.


Subject(s)
AMP-Activated Protein Kinases , Neoplasms , Protein Processing, Post-Translational , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Liver/metabolism , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Neoplasms/enzymology
2.
Int J Mol Sci ; 22(9)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33924999

ABSTRACT

Pancreatic cancer (PC) is the seventh leading cause of cancer death worldwide, and remains one of our most recalcitrant and dismal diseases. In contrast to many other malignancies, there has not been a significant improvement in patient survival over the past decade. Despite advances in our understanding of the genetic alterations associated with this disease, an incomplete understanding of the underlying biology and lack of suitable animal models have hampered efforts to develop more effective therapies. LKB1 is a tumor suppressor that functions as a primary upstream kinase of adenine monophosphate-activated protein kinase (AMPK), which is an important mediator in the regulation of cell growth and epithelial polarity pathways. LKB1 is mutated in a significant number of Peutz-Jeghers syndrome (PJS) patients and in a small proportion of sporadic cancers, including PC; however, little is known about how LKB1 loss contributes to PC development. Here, we report that a reduction in Wnt/ß-catenin activity is associated with LKB1 tumor-suppressive properties in PC. Remarkably, in vivo functional analyses of ß-catenin in the Pdx-1-Cre LKB1L/L ß-cateninL/L mouse model compared to LKB1 loss-driven cystadenoma demonstrate that the loss of ß-catenin impairs cystadenoma development in the pancreas of Pdx-1Cre LKB1L/L mice and dramatically restores the normal development and functions of the pancreas. This study further determined the in vivo and in vitro therapeutic efficacy of the ß-catenin inhibitor FH535 in suppressing LKB1 loss-driven cystadenoma and reducing PC progression that delineates the potential roles of Wnt/ß-catenin signaling in PC harboring LKB1 deficiency.


Subject(s)
Cystadenoma, Mucinous/metabolism , Pancreatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/deficiency , Sulfonamides/pharmacology , beta Catenin/antagonists & inhibitors , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cystadenoma, Mucinous/etiology , Cystadenoma, Mucinous/prevention & control , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/prevention & control , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Protein Serine-Threonine Kinases/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
3.
Mol Metab ; 44: 101131, 2021 02.
Article in English | MEDLINE | ID: mdl-33278637

ABSTRACT

BACKGROUND: Live kinase B1 (LKB1) is a tumor suppressor that is mutated in Peutz-Jeghers syndrome (PJS) and a variety of cancers. Lkb1 encodes serine-threonine kinase (STK) 11 that activates AMP-activated protein kinase (AMPK) and its 13 superfamily members, regulating multiple biological processes, such as cell polarity, cell cycle arrest, embryo development, apoptosis, and bioenergetics metabolism. Increasing evidence has highlighted that deficiency of LKB1 in cancer cells induces extensive metabolic alterations that promote tumorigenesis and development. LKB1 also participates in the maintenance of phenotypes and functions of normal cells through metabolic regulation. SCOPE OF REVIEW: Given the important role of LKB1 in metabolic regulation, we provide an overview of the association of metabolic alterations in glycolysis, aerobic oxidation, the pentose phosphate pathway (PPP), gluconeogenesis, glutamine, lipid, and serine induced by aberrant LKB1 signals in tumor progression, non-neoplastic diseases, and functions of immune cells. MAJOR CONCLUSIONS: In this review, we summarize layers of evidence demonstrating that disordered metabolisms in glucose, glutamine, lipid, and serine caused by LKB1 deficiency promote carcinogenesis and non-neoplastic diseases. The metabolic reprogramming resulting from the loss of LKB1 confers cancer cells with growth or survival advantages. Nevertheless, it also causes a metabolic frangibility for LKB1-deficient cancer cells. The metabolic regulation of LKB1 also plays a vital role in maintaining cellular phenotype in the progression of non-neoplastic diseases. In addition, lipid metabolic regulation of LKB1 plays an important role in controlling the function, activity, proliferation, and differentiation of several types of immune cells. We conclude that in-depth knowledge of metabolic pathways regulated by LKB1 is conducive to identifying therapeutic targets and developing drug combinations to treat cancers and metabolic diseases and achieve immunoregulation.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/physiology , Energy Metabolism/physiology , Glucose/metabolism , Glutamine/metabolism , Humans , Lipid Metabolism , Lipids , Metabolic Engineering , Metabolic Networks and Pathways/genetics , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Serine/metabolism , Signal Transduction
4.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
5.
Turk Patoloji Derg ; 35(3): 247-253, 2019.
Article in English | MEDLINE | ID: mdl-28832082

ABSTRACT

We describe an unusual case of a Peutz-Jeghers syndrome associated with a composite synchronous cervical neoplasia comprising precursor "lobular endocervical glandular hyperplasia (LEGH)", "minimal deviation adenocarcinoma (MDA)" and "gastric-type adenocarcinoma (GTA)" along with a serous tubal intraepithelial lesion (STIL) in the right fallopian tube. A 24-year-old woman presented with a white mucoid discharge and bleeding per vaginum for one year. Histopathological evaluation showed MDA & GTA in FIGO grade III with pelvic lymph node metastasis despite a deceptively bland tumour morphology and low Ki-67 index, indicating an aggressive tumour course and poor prognosis. Diagnostic marker profile in the cervix showed gastric type mucin and positive expression of CK-7, CK-20 (patchy), CEA, and negative CDX-2, p16, ER and PR. Further an attempt at eliciting the oncogenesis pathway in view of the p16 and HPV negative nature of the gastric type cervical adenocarcinoma showed negativity for p53 but activation of cyclin D1. Growth factors including Her2 and EGFR were negative while VEGFR was over-expressed. She was treated by radical hysterectomy and pelvic radiation. She was free from recurrence at the 12-month follow-up. This is a first-time report of a STIL in the fallopian tube which was validated by a unilateral mutant type p53 expression and increased Ki67 index, associated with synchronous gastric type adenocarcinoma of the cervix in all stages of evolution.


Subject(s)
Adenocarcinoma/pathology , Endometrial Hyperplasia/pathology , Fallopian Tube Neoplasms/pathology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Multiple Primary/pathology , Peutz-Jeghers Syndrome/pathology , Precancerous Conditions/pathology , Uterine Cervical Neoplasms/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/therapy , Fallopian Tube Neoplasms/chemistry , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/therapy , Female , Humans , Hysterectomy , Immunohistochemistry , Magnetic Resonance Imaging , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/therapy , Neoplasms, Multiple Primary/chemistry , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/therapy , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/therapy , Radiotherapy, Adjuvant , Tomography, X-Ray Computed , Treatment Outcome , Uterine Cervical Neoplasms/chemistry , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/therapy , Young Adult
6.
Dan Med J ; 63(8)2016 Aug.
Article in English | MEDLINE | ID: mdl-27477802

ABSTRACT

Hamartomatous polyps (HPs) in the gastrointestinal (GI) tract are rare compared to other types of GI polyps, yet they are the most common type of polyp in children. The symptoms are usually rectal bleeding, abdominal pain, obstipation, anaemia, and/or small bowel obstruction. The polyps are typically removed concurrently with endoscopy when located in the colon, rectum, or stomach, whereas polyps in the small bowel are removed during push-enteroscopy, device-assisted enteroscopy, or by surgery. HPs can be classified as juvenile polyps or Peutz-Jeghers polyps based on their histopathological appearance. Patients with one or a few juvenile polyps are usually not offered clinical follow-up as the polyp(s) are considered not to harbour any malignant potential. Nevertheless, it is important to note that juvenile polyps and HPs are also found in patients with hereditary hamartomatous polyposis syndromes (HPS). Patients with HPS have an increased risk of cancer, recurrences of polyps, and extraintestinal complications. The syndromes are important to diagnose, as patients should be offered surveillance from childhood or early adolescence. The syndromes include juvenile polyposis syndrome, Peutz-Jeghers syndrome, and the PTEN hamartoma tumour syndrome. Currently, the HPS diagnoses are based on clinical criteria and are often assisted with genetic testing as candidate genes have been described for each syndrome. This thesis is based on six scientific papers. The overall aim of the studies was to expand the knowledge on clinical course and molecular genetics in patients with HPs and HPS, and to investigate research participants' attitude towards the results of extensive genetic testing.   Paper I: In the first paper we investigated the occurrence, anatomic distribution, and other demographics of juvenile polyps in the colon and rectum in Denmark in 1995-2014. Based on the Danish Pathology Data Bank we found that 1772 patients had 2108 JPs examined in the period, and we calculated the incidence of juvenile polyps to be between 1:45,000 and 1:65,000. The majority of patients with juvenile polyps were adults and 1% fulfilled to diagnostic criteria of JPS. The majority of patients had a single juvenile polyp. Paper II: In this paper we conducted a review of the HPS based on the current literature. Paper III: We investigated the hypothesis that patients with one or few HPs may have a HPS based on genetic screening. We de-signed a panel of 26 genes associated with HPS and used targeted next generation sequencing in 77 patients with mainly one juvenile polyp. We detected several germ line variants, among them three in ENG, two in BMPR1A, one in PTEN, and one in SMAD4. Although some of the detected variants have been reported previously none could be classified as definitely pathogenic or likely pathogenic according to our variant classification scheme and thus we concluded that genetic screening of patients with one or few JPs are not indicated. Paper IV: In Paper IV we investigated one of the ethical aspects of next generation sequencing: the issue whether research participants in NGS studies should be offered the possibility of not re-ceiving information on incidental genetic findings (the "opting out possibility"). We conducted semi-structures interviews in 127 research participants, and found that the majority (61%) wanted information on all incidentals findings, while 36% wanted information on actionable incidental findings. Only 3% did not want information on incidental findings at all. Paper V: In this paper we wanted to gather information on all Danish patients with Peutz-Jeghers syndrome in order to investigate the phenotype and genotype. Through Danish registers we detected 43 patients of which 14 had deceased. We calculated the prevalence of Peutz-Jeghers syndrome to be approximately one in 195,000 individuals. The median age at diagnosis was 29 years with obstruction of the small bowel as the most frequent presenting symptom. We noted 18 cancer occurrences in the population in both the GI tract and at extraintestinal sites, demonstrating that these patients are predisposed to cancer at various anatomical sites. The study also underlined the wide phenotypic expression of the syndrome.   Paper VI: In the last paper we identified patients with juvenile polyposis syndrome, who carry a SMAD4 mutation, and described their genotype and phenotype. We especially investigated whether these patients have symptoms of both juvenile polyposis syndrome and hereditary hemorrhagic telangiectasia. We identified 14 Danish patients. Most of these had symptoms of both conditions and one had aortic root dilatation. Thus, this group of patients requires a multidisciplinary follow-up program.


Subject(s)
Diagnostic Imaging/methods , Genetic Testing/methods , Peutz-Jeghers Syndrome , Genotype , Humans , Peutz-Jeghers Syndrome/diagnosis , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Phenotype
7.
Arkh Patol ; 78(2): 10-18, 2016.
Article in Russian | MEDLINE | ID: mdl-27070770

ABSTRACT

UNLABELLED: Familial adenomatous polyposis (FAP) and Peutz-Jeghers syndrome are genetic diseases characterized by gastrointestinal polyps, extraintestinal manifestations, and autosomal dominant inheritance. The carriers of these diseases from early childhood are at risk for neoplasias at different sites, which are symptomatic at various ages. AIM: to study the clinical organ-specific manifestations in patients with FAP and Peutz-Jeghers, genetics update and possibilities of diagnosis, monitoring, and treatment of these diseases. MATERIAL AND METHODS: The authors give the results of their examination and follow-up of children with FAP and Peutz-Jeghers hamartoma-polypous syndrome. In addition, current data from PubMed, Medline (including reviews, original articles and case reports) were used. RESULTS: The main clinical organ-specific signs of multiple tumors in FAP and Peutz-Jeghers syndrome are shown. Data on the assessment of a risk for malignant tumors at various sites in the affected patients and their family members at different ages are provided. Each of these syndromes has a dissimilar genetic foundation. FAP is caused by the germline mutations in the APC gene, Peutz-Jeghers syndrome is by the STK11 gene, which predispose individuals to specifically associated neoplasias and require different follow-up strategies. Information on a phenotype-genotype correlation may serve as a reference point for the possible severity and various manifestations of a disease. An update on the molecular pathogenesis of these diseases is considered. CONCLUSION: Molecular genetic testing of the genes associated with FAP and Peutz-Jeghers syndromes makes it possible to timely recognize family members at high risk, to plan therapeutic strategy and to affect the course of a disease. The joint participation of pediatricians, proctologists, oncologists, morphologists, geneticists, and molecular biologists is essential to timely recognize the carriers of the syndromes and a better prognosis in these patients.


Subject(s)
Adenomatous Polyposis Coli Protein , Adenomatous Polyposis Coli , Mutation , Peutz-Jeghers Syndrome , Protein Serine-Threonine Kinases , AMP-Activated Protein Kinase Kinases , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Female , Humans , Male , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
8.
Hum Pathol ; 50: 153-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26997450

ABSTRACT

The SHH signaling pathway is critical for gastrointestinal development and organic patterning, and dysregulation of SHH pathway molecules has been detected in multiple gastrointestinal neoplasms. This study investigated the role of the SHH signaling pathway in PJS. Expression of SHH, PTCH, and GLI1 was examined by real-time PCR and immunohistochemistry in 20 normal tissues and 75 colorectal lesions (25 PJPs, 25 adenomas, and 25 adenocarcinomas). Expression of SHH, PTCH, and GLI1 mRNA was higher in PJPs than in normal tissue (P < .05) and gradually increased along the PJP-adenoma-adenocarcinoma sequence (P < .05). Immunostaining indicated that SHH expression was present in 60% of PJPs, 72% of adenomas, and 84% of carcinomas, whereas 68% of PJPs, 72% of adenomas, and 88% of carcinomas exhibited cytoplasmic expression of PTCH. Moreover, high GLI1 expression was detected in 56% of PJPs, 64% of adenomas, and 80% of carcinomas; and high nuclear expression of GLI1 was observed in 8 adenomas with atypia and 15 carcinomas. Increased SHH, PTCH, and GLI1 protein correlated positively with tumor grade (P = .012, P = .003, and P = .007, respectively), tumor depth (P = .024, P = .007, and P = .01), and lymph node metastasis (P = .05, P = .015, and P = .005). This study identified aberrant expression of SHH pathway molecules in PJS, and the findings may supply a novel mechanism for the development of PJ polyps.


Subject(s)
Adenocarcinoma/chemistry , Adenoma/chemistry , Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Hedgehog Proteins/analysis , Peutz-Jeghers Syndrome/metabolism , Receptors, Cell Surface/analysis , Signal Transduction , Transcription Factors/analysis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling/methods , Hedgehog Proteins/genetics , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Patched Receptors , Patched-1 Receptor , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/pathology , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Up-Regulation , Young Adult , Zinc Finger Protein GLI1
9.
Hum Pathol ; 49: 39-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26826408

ABSTRACT

The morphologic features of the gastrointestinal polyps in hamartomatous polyposis syndromes are poorly defined. Our aim was to better characterize the gastrointestinal hamartomas in these syndromes. A blinded review was performed regarding many histologic features for every polyp. The study included 15 Cowden syndrome, 13 Peutz-Jeghers (PJS), 12 juvenile polyposis (JuvPS) patients, and 32 cases of sporadic hamartomatous polyps. A total of 375 polyps were examined. Cowden syndrome polyps were characteristically colonic, sessile, small, without surface erosion, and showing mildly inflamed fibrotic lamina propria with smooth muscle proliferation and lymphoid follicles. They showed the least degree of cystic glands and had no thick mucin. Uncommon but specific features were ganglion cells and nerve fibers within the lamina propria and mucosal fat. PJS polyps were typically of small or large bowel origin, often exophytic, seldom eroded, with inflamed edematous and fibrotic lamina propria and dilated cystic glands filled with often thick mucin. All PJS polyps showed smooth muscle proliferation, frequently widespread. The polyps of JuvPS were typically colonic, large, exophytic, eroded, with strikingly edematous, fibrotic markedly inflamed lamina propria, cystic glands filled with frequently thick mucin, and the least degree of smooth muscle proliferation. Nonsyndromic hamartomatous polyps were similar to JuvPS polyps; however, they were more often colonic, were smaller, showed more widespread smooth muscle proliferation, and were less likely to contain thick mucin. In conclusion, we were able to define the characteristic hamartomatous polyp for each hamartomatous polyposis syndrome. Awareness to these features may aid in the diagnosis of these rare syndromes.


Subject(s)
Colonic Polyps/pathology , Hamartoma Syndrome, Multiple/pathology , Intestinal Polyposis/congenital , Neoplastic Syndromes, Hereditary/pathology , Peutz-Jeghers Syndrome/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Cell Proliferation , Colonic Polyps/chemistry , Diagnosis, Differential , Genetic Testing , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/metabolism , Humans , Intestinal Polyposis/genetics , Intestinal Polyposis/metabolism , Intestinal Polyposis/pathology , Mucins/analysis , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/metabolism , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Predictive Value of Tests , Registries , Utah
10.
Pathol Int ; 64(6): 283-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24965111

ABSTRACT

In this report, unique endocervical glandular lesions exhibiting gastric differentiation were examined in a patient with Peutz-Jeghers syndrome. The result of the human papillomavirus (HPV) in situ hybridization (ISH) for the hysterectomy specimens was negative, but they demonstrated a papillary mucinous adenocarcinoma at the proximal endocervix continuous to atypical lobular endocervical glandular hyperplasia. Both contained MUC6-positive neutral mucin in cytoplasm, and showed different immunoreactivity to p16, Ki-67, and p53. Moreover, they harbored the identical K-RAS gene mutation suggesting that there was a common origin. Somatic K-RAS mutation and defective function of p16 may have been involved in the tumorigenesis of these unusual mucinous neoplasms.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Neoplasm Proteins/metabolism , Peutz-Jeghers Syndrome/pathology , Proto-Oncogene Proteins/metabolism , Uterine Cervical Neoplasms/pathology , ras Proteins/metabolism , Adenocarcinoma, Mucinous/complications , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Cyclin-Dependent Kinase Inhibitor p16 , Female , Humans , Mutation , Peutz-Jeghers Syndrome/complications , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Uterine Cervical Neoplasms/complications , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Young Adult , ras Proteins/genetics
11.
J Clin Endocrinol Metab ; 98(12): E1979-87, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037887

ABSTRACT

CONTEXT: Peutz-Jeghers syndrome (PJS) is an autosomal-dominant disorder that arises as a consequence of mutations in the STK11 gene that encodes LKB1. PJS males often have estrogen excess manifesting as gynecomastia and advanced bone age. We and others have previously described an increase in testicular aromatase expression in PJS patients. However, the underlying mechanism has not yet been explored. OBJECTIVE: The aim of this study was to characterize the role of LKB1 in regulating the expression of aromatase in boys with PJS via signaling pathways involving AMP-activated protein kinase (AMPK) and cyclic AMP-responsive element binding protein-regulated transcription coactivators (CRTCs). PATIENTS: We studied testicular biopsies from two boys with STK11 mutations: a 13-year-old boy and an unrelated 4-year-old boy with prepubertal gynecomastia and advanced bone age, as well as breast tissue from the 13-year-old boy. RESULTS: Loss of heterozygosity of STK11, measured by the absence of LKB1 immunofluorescence, was observed in Sertoli cells of abnormal cords of testis samples from affected individuals. This was associated with loss of p21 expression and decreased phosphorylation of AMPK, known downstream targets of LKB1, as well as the increased expression of aromatase. Similar results of low LKB1 expression in cells expressing aromatase were observed in the mammary epithelium from one of these individuals. Nuclear expression of the CRTC proteins, potent stimulators of aromatase and known to be inhibited by AMPK, was significantly correlated with aromatase. CONCLUSIONS: Loss of heterozygosity of the STK11 gene leads to an increase in aromatase expression associated with an increase in CRTC nuclear localization, thereby providing a mechanism whereby PJS results in increased endogenous estrogens in affected males.


Subject(s)
Aromatase/biosynthesis , Gynecomastia/etiology , Loss of Heterozygosity , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Testis/enzymology , AMP-Activated Protein Kinase Kinases , Adolescent , Cell Nucleus/metabolism , Cell Nucleus/pathology , Child, Preschool , Humans , Male , Mammary Glands, Human/enzymology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Sertoli Cells/enzymology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Testis/metabolism , Testis/pathology , Transcription Factors/metabolism
12.
Pediatr Blood Cancer ; 60(10): E116-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23677888

ABSTRACT

Mismatch DNA repair (MMR) mRNA expression analysis was performed on a biopsy of oral mucosa melanin pigmentation lesion, a hamartomatous polyp and peripheral blood derived from a 12-year-old child with Peutz-Jeghers Syndrome (PJS). We present a deficient MMR system, in a PJS patient, which demonstrated low mRNA levels of hMSH6 and hPMS2 and an increasing MMR deficiency from the non-dysplastic lesion to hamartomatous polyp of PJS with a high risk of cancer.


Subject(s)
DNA Mismatch Repair , Melanins , Mouth/pathology , Peutz-Jeghers Syndrome/genetics , RNA, Messenger/genetics , Skin Pigmentation , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Child , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Mismatch Repair Endonuclease PMS2 , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , RNA, Messenger/biosynthesis
13.
Mod Pathol ; 26(9): 1235-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23599156

ABSTRACT

Peutz-Jeghers syndrome is an autosomal dominant condition characterized by gastrointestinal hamartomatous polyps. The pathologic identification of a Peutz-Jeghers polyp is integral to the diagnosis of this syndrome that often remains undiagnosed until after these polyps are identified. Histologically, Peutz-Jeghers polyps are characterized by a distinctive arborization of smooth muscle within the lamina propria. Colonic Peutz-Jeghers polyps, however, may mimic mucosal prolapse polyps or virtually any colonic polyp that undergoes prolapse. In this paper, we explore the morphological features of colonic Peutz-Jeghers polyps and the diagnostic challenges associated with these polyps. Colonic polyps from patients with Peutz-Jeghers syndrome were identified (n=34). The control cohort, included mucosal prolapse polyps (n=5), hyperplastic polyps (n=10) and tubular adenomas with prolapse (n=9), ganglioneuromatous polyps (n=2) and juvenile polyps (n=14). Intramucosal smooth muscle fibers were identified in all classes of polyps. Twenty-three of the 34 colonic Peutz-Jeghers polyps were characterized by lobulated clusters of colonic crypts. On immunohistochemistry, desmin-positive smooth muscle fibers were seen surrounding these lobules. This lobular organization of the crypts was not identified in mucosal prolapse polyps and hyperplastic polyps or tubular adenomas with prolapse; only one of the 14 juvenile polyps showed this pattern of reactivity on a desmin stain. Our data suggests that the histologic hallmark of colonic Peutz-Jeghers polyps is the lobular organization of the crypts, and that an arborizing pattern of smooth muscle proliferation is neither sensitive nor a specific marker of colonic Peutz-Jeghers polyps. The presence of desmin-positive smooth muscle fibers surrounding the lobules is a helpful diagnostic feature of colonic Peutz-Jeghers polyps, and facilitates the distinction of these polyps from non-Peutz-Jeghers polyps with prolapse-like changes.


Subject(s)
Colonic Neoplasms/pathology , Colonic Polyps/pathology , Peutz-Jeghers Syndrome/pathology , Adult , Biomarkers, Tumor/analysis , Case-Control Studies , Colonic Neoplasms/chemistry , Colonic Polyps/chemistry , Desmin/analysis , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Intestine, Small/chemistry , Intestine, Small/pathology , Male , Muscle, Smooth/chemistry , Muscle, Smooth/pathology , Peutz-Jeghers Syndrome/metabolism , Predictive Value of Tests , Young Adult
14.
Gut ; 61(2): 202-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21813469

ABSTRACT

BACKGROUND AND AIMS: LKB1 is a serine-threonine kinase, mutation of which can lead to the development of multiple benign intestinal hamartomas (Peutz-Jeghers syndrome). In this study, the authors investigate the mechanisms underlying this phenotype by exploring the transcriptional changes associated with Lkb1 deletion in intestinal epithelium. METHODS: The authors used mice with Lkb1 deleted in the intestinal epithelium using a Cyp1a1-specific inducible Cre recombinase and used Affymetrix (Santa Clara, California, USA) microarray analysis to examine the transcriptional changes occurring immediately after Lkb1 loss. The authors also generated crypt-villus organoid culture to analyse Lkb1 role in intestinal responses to exogenous stimuli. RESULTS: Affymetrix analysis identified the most significant change to be in Ren1 expression, a gene encoding a protease involved in angiotensinogen processing. Lkb1 deletion also enhanced ACE expression and subsequently angiotensin II (AngII) production in the mouse intestine. Intestinal apoptosis induced by Lkb1 deficiency was suppressed by ACE inhibitor captopril. Lkb1-deficient intestinal epithelium showed dynamic changes in AngII receptor type 1, suggesting a possible compensatory response to elevated AngII levels. A similar reduction in epithelial AngII receptor type 1 was also observed in human Peutz-Jeghers syndrome tumours contrasting with high expression of the receptor in the tumour stroma. Mechanistically, the authors showed two pieces of data that position Lkb1 in renin expression regulation, and they implied the importance of Lkb1 in linking cell responses with nutrient levels. First, the authors showed that Lkb1 deletion in isolated epithelial organoid culture resulted in renin upregulation only when the organoids were challenged with external cues such as AngII; second, that renin upregulation was dependent upon the MEK/ERK pathway in a circadian fashion and corresponded to active feeding time when nutrient levels were high. CONCLUSIONS: Taken together, these data reveal a novel role for Lkb1 in regulation of the gastrointestinal renin-angiotensin system.


Subject(s)
Angiotensin II/metabolism , Gene Deletion , Intestinal Mucosa/metabolism , Peptidyl-Dipeptidase A/metabolism , Protein Serine-Threonine Kinases/genetics , Renin-Angiotensin System/physiology , Renin/metabolism , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Mucin-2/metabolism , Oligonucleotide Array Sequence Analysis , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/metabolism , Protein Serine-Threonine Kinases/metabolism , Renin/genetics , Renin-Angiotensin System/genetics
15.
Nat Rev Endocrinol ; 8(3): 135-47, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22024975

ABSTRACT

Aromatase, an enzyme located in the endoplasmic reticulum of estrogen-producing cells, catalyzes the rate-limiting step in the conversion of androgens to estrogens in many tissues. The clinical features of patients with defects in CYP19A1, the gene encoding aromatase, have revealed a major role for this enzyme in epiphyseal plate closure, which has promoted interest in the use of inhibitors of aromatase to improve adult height. The availability of the selective aromatase inhibitors letrozole and anastrozole--currently approved as adjuvant therapy for breast cancer--have stimulated off-label use of aromatase inhibitors in pediatrics for the following conditions: hyperestrogenism, such as aromatase excess syndrome, Peutz-Jeghers syndrome, McCune-Albright syndrome and functional follicular ovarian cysts; hyperandrogenism, for example, testotoxicosis (also known as familial male-limited precocious puberty) and congenital adrenal hyperplasia; pubertal gynecomastia; and short stature and/or pubertal delay in boys. Current data suggest that aromatase inhibitors are probably effective in the treatment of patients with aromatase excess syndrome or testotoxicosis, partially effective in Peutz-Jeghers and McCune-Albright syndrome, but probably ineffective in gynecomastia. Insufficient data are available in patients with congenital adrenal hyperplasia or functional ovarian cysts. Although aromatase inhibitors appear effective in increasing adult height of boys with short stature and/or pubertal delay, safety concerns, including vertebral deformities, a decrease in serum HDL cholesterol levels and increase of erythrocytosis, are reasons for caution.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Body Height/drug effects , Child Development/drug effects , Age Factors , Animals , Aromatase Inhibitors/adverse effects , Body Height/physiology , Child , Child Development/physiology , Female , Humans , Hyperandrogenism/drug therapy , Hyperandrogenism/metabolism , Male , Peutz-Jeghers Syndrome/drug therapy , Peutz-Jeghers Syndrome/metabolism
16.
Fam Cancer ; 10(3): 469-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21826537

ABSTRACT

Peutz-Jeghers patients frequently develop clinically significant complications, namely hemorrhage and bowel obstruction, from small intestinal hamartomatous polyps that frequently require surgery. In addition, many PJS patients develop epithelial malignancies in a variety of organs. The vast majority of PJS is due to germline alterations in the STK11 gene that encodes a protein that modulates PI3-kinase signaling, a key regulator of cell survival and growth. One of the major downstream mediators of PI3-kinase signaling is mTOR, the mammalian target of rapamycin. Several drugs that inhibit the PI3-kinase signal transduction pathway are in development and one, RAD001 (everolimus), an mTOR inhibitor, was recently approved for the treatment of renal cell carcinoma. Effective chemoprevention of intestinal polyps would be a first step in simplifying and improving the management of PJS patients. We present here, the rationale for the first human PJS chemoprevention trial using an mTOR inhibitor.


Subject(s)
Antineoplastic Agents/therapeutic use , Peutz-Jeghers Syndrome/prevention & control , TOR Serine-Threonine Kinases/antagonists & inhibitors , Clinical Trials as Topic , Humans , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
17.
Fam Cancer ; 10(3): 437-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21805166

ABSTRACT

Familial cancer syndromes present rare insights into malignant tumor development. The molecular background of polyp formation and the cancer prone state in Peutz-Jeghers syndrome remain enigmatic to this day. Previously, we proposed that Peutz-Jeghers polyps are not pre-malignant lesions, but an epiphenomenon to the malignant condition. However, Peutz-Jeghers polyp formation and the cancer-prone state must both be accounted for by the same molecular mechanism. Our contribution focuses on the histopathology of the characteristic Peutz-Jeghers polyp and recent research on stem cell dynamics and how these concepts relate to Peutz-Jeghers polyposis. We discuss a protracted clonal evolution scenario in Peutz-Jeghers syndrome due to a germline LKB1 mutation. Peutz-Jeghers polyp formation and malignant transformation are separately mediated through the same molecular mechanism played out on different timescales. Thus, a single mechanism accounts for the development of benign Peutz-Jeghers polyps and for malignant transformation in Peutz-Jeghers syndrome.


Subject(s)
Aberrant Crypt Foci/metabolism , Aberrant Crypt Foci/pathology , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Humans
18.
Fam Cancer ; 10(3): 425-35, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21519908

ABSTRACT

The Peutz-Jeghers syndrome (PJS) culprit kinase LKB1 phosphorylates and activates multiple intracellular kinases regulating cell metabolism and polarity. The relevance of each of these pathways is highly variable depending on the tissue type, but typically represents functions of differentiated cells. These include formation and maintenance of specialized cell compartments in nerve axons, swift refunneling of metabolites and restructuring of cell architecture in response to environmental cues in committed lymphocytes, and ensuring energy-efficient oxygen-based energy expenditure. Such features are often lost or reduced in cancer cells, and indeed LKB1 defects in PJS-associated and sporadic cancers and even the benign PJS polyps lead to differentiation defects, including expansion of partially differentiated epithelial cells in PJS polyps and epithelial-to-mesenchymal transition in carcinomas. This review focuses on the involvement of LKB1 in the differentiation of epithelial, mesenchymal, hematopoietic and germinal lineages.


Subject(s)
Cell Differentiation , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Cell Polarity , Humans
19.
J Pathol ; 223(5): 584-92, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21341271

ABSTRACT

Peutz-Jeghers syndrome (PJS) is a rare, inherited disease caused by germline mutation of the LKB1 gene. Patients with PJS develop characteristic polyps in the digestive tract and carry an elevated risk of cancers in multiple organs, including the intestinal tract. While LKB1 is capable of phosphorylating AMPK and regulates the mTOR pathway, it is also known to be a multitasking protein that can influence other cellular processes, including cell polarity. We hypothesized that there may be other biological pathways directly or indirectly affected by the loss of LKB1 in PJS and aimed to investigate this possibility through transcriptional profiling of polyps harvested from an Lkb1(+/-) mouse model of PJS and from PJS patients. We identified alterations in the mRNA level of a wide range of genes, including some that are involved in Wnt signalling (Wnt5a, Wif1, Dixdc1, Wnt11, Ccnd1, and Ccnd2), although we did not observe nuclear localization of ß-catenin in over 93 human PJS intestinal polyps or in 24 gastric polyps from Lkb1(+/-) mice. Among these genes, WNT5A, a non-canonical and non-transforming Wnt, is consistently up-regulated in both Lkb1(+/-) mice and human PJS polyps at a high level. We performed in situ hybridization to further define the spatial expression pattern of WNT5A and observed a strong signal in the stroma of mouse and human polyps compared to no or very low expression in the mucosa. Our findings indicate that WNT5A plays an important role in PJS polyposis.


Subject(s)
Peutz-Jeghers Syndrome/metabolism , Wnt Proteins/biosynthesis , Animals , Gastric Mucosa/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study/methods , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis/methods , Peutz-Jeghers Syndrome/genetics , Polyps/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Signal Transduction , Stomach Diseases/metabolism , Wnt Proteins/genetics , Wnt Proteins/physiology , Wnt-5a Protein
20.
Oncol Rep ; 23(6): 1569-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428811

ABSTRACT

The premalignant potential of Peutz-Jeghers syndrome (PJS) hamartomas has not been established. The major gene responsible for PJS is LKB1. LKB1 has a complex cellular role, therefore, the exact role of LKB1 in Peutz-Jeghers syndrome hamartomas (PJSs) is particularly difficult to understand. It has recently been found that LKB1 functions in the Wnt pathway in Xenopus during early development. Aberrant beta-catenin expression, the key regulator of the activated Wnt/beta-catenin signaling pathway, appears to stimulate interferon-induced gene 1 (IFITM1) products in intestinal tumorigenesis. Both contribute to intestinal tumor formation and tumor progression. This study was designed to investigate expression of LKB1, beta-catenin and IFITM1 in PJSs, colorectal adenomas (CRAs), colorectal carcinomas (CRCs) and normal colorectal mucosas (NCs) using RT-PCR and immunohistochemistry. Immunofluorescence was used to assess the co-expression characteristics of beta-catenin and IFITM1. Results showed that the expression profiles of LKB1, beta-catenin and IFITM1 in PJSs were similar to those in CRAs both at the mRNA and protein levels. The cytoplasmic level of beta-catenin expression correlated strongly with LKB1 and IFITM1 expression in the tumor cells. The dyregulation of beta-catenin was found in a majority (16/20) of the PJS polyps. Immunofluorescence also revealed co-expression of beta-catenin and IFITM1 in the cytoplasm of the PJSs. These findings suggest that Wnt signaling may be activated in a subset of PJSs, and activation of the Wnt/beta-catenin signaling in PJS polyps may be caused by LKB1 expression. The activated beta-catenin signaling pathway including IFITM1 might play an important role in a subset of PJS polyps.


Subject(s)
Adenoma/metabolism , Colorectal Neoplasms/metabolism , Membrane Proteins/metabolism , Peutz-Jeghers Syndrome/metabolism , Protein Serine-Threonine Kinases/metabolism , beta Catenin/metabolism , AMP-Activated Protein Kinase Kinases , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Adolescent , Adult , Aged , Antigens, Differentiation , Blotting, Western , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Male , Membrane Proteins/genetics , Middle Aged , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Rectum/metabolism , Rectum/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Young Adult , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...