Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Endocr Rev ; 40(6): 1468-1480, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31081903

ABSTRACT

PTH is an important regulator of calcium and phosphate homeostasis and bone remodeling. It is metabolized into PTH fragments, which are measured to a different extent by PTH assays of different generations because of differences in fragments recognized and lack of assay standardization. PTH is measured in the workup of several conditions, and clinical guidelines provide recommendations concerning these measurements. This review provides an overview of the impact of differences between PTH assays, applying distinct clinical guidelines for primary and secondary hyperparathyroidism and perioperative use of PTH measurements. Guidelines deal with PTH measurement in different ways, recommending either trend monitoring, the use of a fold increase of the upper reference limit, or an absolute PTH cutoff value. For classic primary hyperparathyroidism (PHPT), the type of PTH assay used will not affect diagnosis or management because the precise concentration of PTH is less relevant. In chronic kidney disease, the guideline recommends treating secondary hyperparathyroidism above a twofold to ninefold PTH increase, which will result in different clinical decisions depending on the assay used. For patients after bariatric surgery, guidelines state absolute cutoff values for PTH, but the impact of different generation assays is unknown because direct comparison of PTH assays has never been performed. During parathyroid surgery, PTH measurements with a third-generation assay reflect treatment success more rapidly than second-generation assays. Increased awareness among clinicians regarding the complexity of PTH measurements is warranted because it can affect clinical decisions.


Subject(s)
Parathyroid Hormone/blood , Calcium/blood , Humans , Hyperparathyroidism/blood , Hyperparathyroidism/diagnosis , Immunoassay , Mass Spectrometry
2.
Cell Rep ; 25(11): 3021-3035.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540936

ABSTRACT

Leukemic stem cells (LSCs) are thought to be the major cause of the recurrence of acute myeloid leukemia (AML) due to their potential for self-renewal. To identify therapeutic strategies targeting LSCs, while sparing healthy hematopoietic stem cells (HSCs), we performed gene expression profiling of LSCs, HSCs, and leukemic progenitors all residing within the same AML bone marrow and identified insulin-like growth factor-binding protein 7 (IGFBP7) as differentially expressed. Low IGFBP7 is a feature of LSCs and is associated with reduced chemotherapy sensitivity. Enhancing IGFBP7 by overexpression or addition of recombinant human IGFBP7 (rhIGFBP7) resulted in differentiation, inhibition of cell survival, and increased chemotherapy sensitivity of primary AML cells. Adding rhIGFBP7 reduced leukemic stem and/or progenitor survival and reversed a stem-like gene signature, but it had no influence on normal hematopoietic stem cell survival. Our data suggest a potential clinical utility of the addition of rhIGFBP7 to current chemotherapy regimens to decrease AML relapse rates.


Subject(s)
Cell Differentiation , Hematopoiesis , Insulin-Like Growth Factor Binding Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Bone Marrow/pathology , Cell Differentiation/drug effects , Cell Survival/drug effects , Clone Cells , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Recombinant Proteins/pharmacology
3.
Blood ; 127(4): 458-63, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26582376

ABSTRACT

Enhanced expression of ecotropic viral integration site 1 (EVI-1) occurs in ∼10% of acute myeloid leukemia (AML) patients and is associated with a very poor disease outcome. Patients with EVI-1-positive AML have poor initial responses to chemotherapy and high relapse rates, indicating an urgent need for alternative treatment strategies improving clinical outcome for these patients. Because treatment of acute promyelocytic patients with all-trans retinoic acid (ATRA) has improved the survival of these patients substantially, we investigated whether ATRA might also be effective for the subgroup of AML patients with EVI-1 overexpression. Here, we show that a substantial part of the EVI-1-positive AML cases respond to ATRA by induction of differentiation and decreased clonogenic capacity of myeloid blasts. Most importantly, we demonstrate that in vivo treatment of primary EVI-1-positive AML with ATRA leads to a significant reduction in leukemic engraftment. Altogether, our results show that a considerable part of the EVI-1-positive primary AML cases are sensitive to ATRA, suggesting that combining ATRA with the currently used conventional chemotherapy might be a promising treatment strategy decreasing relapse rates and enhancing complete remissions in this poor prognostic subgroup of AML patients.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogenes/genetics , Transcription Factors/genetics , Tretinoin/pharmacology , Animals , Cell Differentiation/drug effects , DNA-Binding Proteins/analysis , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MDS1 and EVI1 Complex Locus Protein , Male , Mice, SCID , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/pathology , Transcription Factors/analysis , Tumor Cells, Cultured , Up-Regulation
4.
Pigment Cell Melanoma Res ; 28(3): 307-17, 2015 May.
Article in English | MEDLINE | ID: mdl-25728708

ABSTRACT

No effective targeted therapy is currently available for NRAS mutant melanoma. Experimental MEK inhibition is rather toxic and has only limited efficacy in clinical trials. At least in part, this is caused by the emergence of drug resistance, which is commonly seen for single agent treatment and shortens clinical responses. Therefore, there is a dire need to identify effective companion drug targets for NRAS mutant melanoma. Here, we show that at concentrations where single drugs had little effect, ROCK inhibitors GSK269962A or Fasudil, in combination with either MEK inhibitor GSK1120212 (Trametinib) or ERK inhibitor SCH772984 cooperatively caused proliferation inhibition and cell death in vitro. Simultaneous inhibition of MEK and ROCK caused induction of BimEL , PARP, and Puma, and hence apoptosis. In vivo, MEK and ROCK inhibition suppressed growth of established tumors. Our findings warrant clinical investigation of the effectiveness of combinatorial targeting of MAPK/ERK and ROCK in NRAS mutant melanoma.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , rho-Associated Kinases/antagonists & inhibitors , Cell Line, Tumor , Humans , Melanoma/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , rho-Associated Kinases/metabolism
5.
Mol Syst Biol ; 10: 772, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25538140

ABSTRACT

Treatment of BRAF mutant melanomas with specific BRAF inhibitors leads to tumor remission. However, most patients eventually relapse due to drug resistance. Therefore, we designed an integrated strategy using (phospho)proteomic and functional genomic platforms to identify drug targets whose inhibition sensitizes melanoma cells to BRAF inhibition. We found many proteins to be induced upon PLX4720 (BRAF inhibitor) treatment that are known to be involved in BRAF inhibitor resistance, including FOXD3 and ErbB3. Several proteins were down-regulated, including Rnd3, a negative regulator of ROCK1 kinase. For our genomic approach, we performed two parallel shRNA screens using a kinome library to identify genes whose inhibition sensitizes to BRAF or ERK inhibitor treatment. By integrating our functional genomic and (phospho)proteomic data, we identified ROCK1 as a potential drug target for BRAF mutant melanoma. ROCK1 silencing increased melanoma cell elimination when combined with BRAF or ERK inhibitor treatment. Translating this to a preclinical setting, a ROCK inhibitor showed augmented melanoma cell death upon BRAF or ERK inhibition in vitro. These data merit exploration of ROCK1 as a target in combination with current BRAF mutant melanoma therapies.


Subject(s)
Melanoma/genetics , Proto-Oncogene Proteins B-raf/metabolism , rho-Associated Kinases/metabolism , Cell Line, Tumor , Chromatography, Liquid , Down-Regulation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Indazoles/pharmacology , Indoles/pharmacology , Molecular Targeted Therapy , Mutation , Piperazines/pharmacology , Proteomics , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Sulfonamides/pharmacology , Tandem Mass Spectrometry , Vemurafenib , rho-Associated Kinases/genetics
6.
Cell Rep ; 9(4): 1375-86, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456132

ABSTRACT

To identify factors preferentially necessary for driving tumor expansion, we performed parallel in vitro and in vivo negative-selection short hairpin RNA (shRNA) screens. Melanoma cells harboring shRNAs targeting several DNA damage response (DDR) kinases had a greater selective disadvantage in vivo than in vitro, indicating an essential contribution of these factors during tumor expansion. In growing tumors, DDR kinases were activated following hypoxia. Correspondingly, depletion or pharmacologic inhibition of DDR kinases was toxic to melanoma cells, including those that were resistant to BRAF inhibitor, and this could be enhanced by angiogenesis blockade. These results reveal that hypoxia sensitizes melanomas to targeted inhibition of the DDR and illustrate the utility of in vivo shRNA dropout screens for the identification of pharmacologically tractable targets.


Subject(s)
DNA Damage , DNA Repair , Genetic Testing , Melanoma/genetics , Melanoma/pathology , RNA Interference , Animals , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2/metabolism , DNA Repair/drug effects , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Stability/drug effects , RNA Interference/drug effects , RNA, Small Interfering/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Pigment Cell Melanoma Res ; 26(4): 542-54, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23590314

ABSTRACT

The involvement of epigenetic alterations in the pathogenesis of melanoma is increasingly recognized. Here, we performed genome-wide DNA methylation analysis of primary cutaneous melanoma and benign melanocytic nevus interrogating 14 495 genes using BeadChip technology. This genome-wide view of promoter methylation in primary cutaneous melanoma revealed an array of recurrent DNA methylation alterations with potential diagnostic applications. Among 106 frequently hypermethylated genes, there were many novel methylation targets and tumor suppressor genes. Highly recurrent methylation of the HOXA9, MAPK13, CDH11, PLEKHG6, PPP1R3C, and CLDN11 genes was established. Promoter methylation of MAPK13, encoding p38δ, was present in 67% of primary and 85% of metastatic melanomas. Restoration of MAPK13 expression in melanoma cells exhibiting epigenetic silencing of this gene reduced proliferation, indicative of tumor suppressive functions. This study demonstrates that DNA methylation alterations are widespread in melanoma and suggests that epigenetic silencing of MAPK13 contributes to melanoma progression.


Subject(s)
Epigenesis, Genetic , Melanoma/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Promoter Regions, Genetic , Skin Neoplasms/metabolism , Cell Proliferation , CpG Islands , DNA Methylation , Disease Progression , Epigenomics , Fibroblasts/cytology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Genome-Wide Association Study , Humans , Melanocytes/cytology , Nevus/metabolism , Sulfites/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism , Melanoma, Cutaneous Malignant
8.
Proc Natl Acad Sci U S A ; 110(13): 5139-44, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23483055

ABSTRACT

Metastasis confronts clinicians with two major challenges: estimating the patient's risk of metastasis and identifying therapeutic targets. Because they are key signal integrators connecting cellular processes to clinical outcome, we aimed to identify transcriptional nodes regulating cancer cell metastasis. Using rodent xenograft models that we previously developed, we identified the transcription factor Fos-related antigen-1 (Fra-1) as a key coordinator of metastasis. Because Fra-1 often is overexpressed in human metastatic breast cancers and has been shown to control their invasive potential in vitro, we aimed to assess the implication and prognostic significance of the Fra-1-dependent genetic program in breast cancer metastasis and to identify potential Fra-1-dependent therapeutic targets. In several in vivo assays in mice, we demonstrate that stable RNAi depletion of Fra-1 from human breast cancer cells strongly suppresses their ability to metastasize. These results support a clinically important role for Fra-1 and the genetic program it controls. We show that a Fra-1-dependent gene-expression signature accurately predicts recurrence of breast cancer. Furthermore, a synthetic lethal drug screen revealed that antagonists of the adenosine receptor A2B (ADORA2B) are preferentially toxic to breast tumor cells expressing Fra-1. Both RNAi silencing and pharmacologic blockade of ADORA2B inhibited filopodia formation and invasive activity of breast cancer cells and correspondingly reduced tumor outgrowth in the lungs. These data show that Fra-1 activity is causally involved in and is a prognostic indicator of breast cancer metastasis. They suggest that Fra-1 activity predicts responsiveness to inhibition of pharmacologically tractable targets, such as ADORA2B, which may be used for clinical interference of metastatic breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Adenosine A2B/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Proto-Oncogene Proteins c-fos/genetics , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/pathology , Rats , Receptor, Adenosine A2B/genetics , Transplantation, Heterologous , Xenograft Model Antitumor Assays
9.
Genes Dev ; 26(10): 1055-69, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22549727

ABSTRACT

Human melanocytic nevi (moles) are benign lesions harboring activated oncogenes, including BRAF. Although this oncogene initially acts mitogenically, eventually, oncogene-induced senescence (OIS) ensues. Nevi can infrequently progress to melanomas, but the mechanistic relationship with OIS is unclear. We show here that PTEN depletion abrogates BRAF(V600E)-induced senescence in human fibroblasts and melanocytes. Correspondingly, in established murine BRAF(V600E)-driven nevi, acute shRNA-mediated depletion of PTEN prompted tumor progression. Furthermore, genetic analysis of laser-guided microdissected human contiguous nevus-melanoma specimens recurrently revealed identical mutations in BRAF or NRAS in adjacent benign and malignant melanocytes. The PI3K pathway was often activated through either decreased PTEN or increased AKT3 expression in melanomas relative to their adjacent nevi. Pharmacologic PI3K inhibition in melanoma cells suppressed proliferation and induced the senescence-associated tumor suppressor p15(INK4B). This treatment also eliminated subpopulations resistant to targeted BRAF(V600E) inhibition. Our findings suggest that a significant proportion of melanomas arise from nevi. Furthermore, these results demonstrate that PI3K pathway activation serves as a rate-limiting event in this setting, acting at least in part by abrogating OIS. The reactivation of senescence features and elimination of cells refractory to BRAF(V600E) inhibition by PI3K inhibition warrants further investigation into the therapeutic potential of simultaneously targeting these pathways in melanoma.


Subject(s)
Cellular Senescence , Melanoma/pathology , Nevus/pathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology , Amino Acid Substitution , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Enzyme Activation , Fibroblasts/metabolism , Fibroblasts/pathology , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Nevus/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism , Valine/genetics , Valine/metabolism
10.
Aging (Albany NY) ; 2(10): 735-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20975209

ABSTRACT

The epithelial-mesenchymal transition is involved in several physiological processes. However, it is also believed to contribute to cancer progression. Conversely, cellular senescence constitutes a failsafe program against cancer progression. Interestingly, EMT and senescence seem to cross paths, with several factors playing dominant roles in both settings. Here, we describe recent observations that link these important cellular processes.


Subject(s)
Cellular Senescence/physiology , Epithelial-Mesenchymal Transition/physiology , Neoplasms/etiology , Animals , Cadherins/metabolism , Catenins/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Homeodomain Proteins/metabolism , Humans , Models, Biological , Neoplasms/metabolism , Oncogene Proteins/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Zinc Finger E-box-Binding Homeobox 1
11.
Mol Cell Biol ; 29(13): 3722-37, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19414595

ABSTRACT

In a genomewide anoikis suppression screen for metastasis genes, we previously identified the neurotrophic receptor tyrosine kinase TrkB. In mouse xenografts, activated TrkB caused highly invasive and metastatic tumors. Here, we describe that TrkB also induces a strong morphological transformation, resembling epithelial-mesenchymal transition (EMT). This required TrkB kinase activity, a functional mitogen-activated protein kinase pathway, suppression of E-cadherin, and induction of Twist, a transcription factor contributing to EMT and metastasis. RNA interference (RNAi)-mediated Twist depletion blocked TrkB-induced EMT-like transformation, anoikis suppression, and growth of tumor xenografts. By searching for essential effectors of TrkB-Twist signaling, we found that Twist induces Snail, another EMT regulator associated with poor cancer prognosis. Snail depletion impaired EMT-like transformation and anoikis suppression induced by TrkB, but in contrast to Twist depletion, it failed to inhibit tumor growth. Instead, Snail RNAi specifically impaired the formation of lung metastases. Epistasis experiments suggested that Twist acts upstream from Snail. Our results demonstrate that TrkB signaling activates a Twist-Snail axis that is critically involved in EMT-like transformation, tumorigenesis, and metastasis. Moreover, our data shed more light on the epistatic relationship between Twist and Snail, two key transcriptional regulators of EMT and metastasis.


Subject(s)
Anoikis/physiology , Epithelium/physiology , Mesoderm/physiology , Neoplasm Metastasis , Receptor, trkB/metabolism , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cadherins/metabolism , Cell Line , Cell Movement/physiology , Epithelial Cells/physiology , Humans , Mesoderm/cytology , Mice , Morphogenesis/physiology , Rats , Receptor, trkB/genetics , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/genetics , Twist-Related Protein 1/genetics
12.
SELECTION OF CITATIONS
SEARCH DETAIL
...