Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Haematologica ; 108(11): 2946-2958, 2023 11 01.
Article in English | MEDLINE | ID: mdl-36951168

ABSTRACT

Drug resistance underpins poor outcomes in many malignancies including refractory and relapsed acute myeloid leukemia (R/R AML). Glucuronidation is a common mechanism of drug inactivation impacting many AML therapies, e.g., cytarabine, decitabine, azacytidine and venetoclax. In AML cells, the capacity for glucuronidation arises from increased production of the UDP-glucuronosyltransferase 1A (UGT1A) enzymes. UGT1A elevation was first observed in AML patients who relapsed after response to ribavirin, a drug used to target the eukaryotic translation initiation factor eIF4E, and subsequently in patients who relapsed on cytarabine. UGT1A elevation resulted from increased expression of the sonic-hedgehog transcription factor GLI1. Vismodegib inhibited GLI1, decreased UGT1A levels, reduced glucuronidation of ribavirin and cytarabine, and re-sensitized cells to these drugs. Here, we examined if UGT1A protein levels, and thus glucuronidation activity, were targetable in humans and if this corresponded to clinical response. We conducted a phase II trial using vismodegib with ribavirin, with or without decitabine, in largely heavily pre-treated patients with high-eIF4E AML. Pre-therapy molecular assessment of patients' blasts indicated highly elevated UGT1A levels relative to healthy volunteers. Among patients with partial response, blast response or prolonged stable disease, vismodegib reduced UGT1A levels, which corresponded to effective targeting of eIF4E by ribavirin. In all, our studies are the first to demonstrate that UGT1A protein, and thus glucuronidation, are targetable in humans. These studies pave the way for the development of therapies that impair glucuronidation, one of the most common drug deactivation modalities. Clinicaltrials.gov: NCT02073838.


Subject(s)
Glucuronosyltransferase , Leukemia, Myeloid, Acute , Humans , Decitabine/therapeutic use , Glucuronosyltransferase/metabolism , Glucuronosyltransferase/therapeutic use , Ribavirin/therapeutic use , Ribavirin/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/therapeutic use , Eukaryotic Initiation Factor-4E/metabolism , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/therapeutic use , Molecular Targeted Therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Cytarabine , Uridine Diphosphate/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
J Immunother Cancer ; 5(1): 83, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29157311

ABSTRACT

BACKGROUND: Checkpoint blockade with ipilimumab provides long-term survival to a significant proportion of patients with metastatic melanoma. New approaches to increase survival and to predict which patients will benefit from treatment are needed. This phase II trial combined ipilimumab with carboplatin/paclitaxel (CP) to assess its safety, efficacy, and to search for peripheral and tumor-based predictive biomarkers. METHODS: Thirty patients with untreated unresectable/metastatic melanoma were treated with ipilimumab and CP. Adverse events (AEs) were monitored and response to treatment was evaluated. Tumor tissue and peripheral blood were collected at specified time points to characterize tumor immune markers by immunohistochemistry and systemic immune activity by multiplex assays and flow cytometry. RESULTS: Eighty three percent of patients received all 5 cycles of CP and 93% completed ipilimumab induction. Serious AEs occurred in 13% of patients, and no treatment-related deaths were observed. Best Overall Response Rate (BORR) and Disease Control Rate (DCR) were 27 and 57%, respectively. Median overall survival was 16.2 months. Response to treatment was positively correlated with a higher tumor CD3+ infiltrate (immune score) at baseline. NRAS and BRAF mutations were less frequent in patients who experienced clinical benefit. Assessment of peripheral blood revealed that non-responders had elevated baseline levels of CXCL8 and CCL4, and a higher proportion of circulating late differentiated B cells. Pre-existing high levels of chemokines (CCL3, CCL4 and CXCL8) and advanced B cell differentiation were strongly associated with worse patient overall survival. Elevated proportions of circulating CD8+/PD-1+ T cells during treatment were associated with worse survival. CONCLUSIONS: The combination of ipilimumab and CP was well tolerated and revealed novel characteristics associated with patients likely to benefit from treatment. A pre-existing systemic inflammatory state characterized by elevation of selected chemokines and advanced B cell differentiation, was strongly associated with poor patient outcomes, revealing potential predictive circulating biomarkers. TRIAL REGISTRATION: Clinicaltrials.gov , NCT01676649 , registered on August 29, 2012.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Ipilimumab/therapeutic use , Melanoma/drug therapy , Paclitaxel/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Female , Humans , Ipilimumab/pharmacology , Male , Melanoma/pathology , Middle Aged , Neoplasm Staging , Paclitaxel/pharmacology , Skin Neoplasms/pathology , Treatment Outcome
5.
Nature ; 511(7507): 90-3, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24870236

ABSTRACT

Drug resistance is a major hurdle in oncology. Responses of acute myeloid leukaemia (AML) patients to cytarabine (Ara-C)-based therapies are often short lived with a median overall survival of months. Therapies are under development to improve outcomes and include targeting the eukaryotic translation initiation factor (eIF4E) with its inhibitor ribavirin. In a Phase II clinical trial in poor prognosis AML, ribavirin monotherapy yielded promising responses including remissions; however, all patients relapsed. Here we identify a novel form of drug resistance to ribavirin and Ara-C. We observe that the sonic hedgehog transcription factor glioma-associated protein 1 (GLI1) and the UDP glucuronosyltransferase (UGT1A) family of enzymes are elevated in resistant cells. UGT1As add glucuronic acid to many drugs, modifying their activity in diverse tissues. GLI1 alone is sufficient to drive UGT1A-dependent glucuronidation of ribavirin and Ara-C, and thus drug resistance. Resistance is overcome by genetic or pharmacological inhibition of GLI1, revealing a potential strategy to overcome drug resistance in some patients.


Subject(s)
Drug Resistance, Neoplasm , Glucuronic Acid/metabolism , Glucuronosyltransferase/metabolism , Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cytarabine/metabolism , Cytarabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Deletion , Glucuronosyltransferase/biosynthesis , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Ribavirin/metabolism , Ribavirin/pharmacology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Zinc Finger Protein GLI1
6.
Front Pharmacol ; 4: 9, 2013.
Article in English | MEDLINE | ID: mdl-23408639

ABSTRACT

Acute myeloid leukemia (AML) with inversion of chromosome 3 is characterized by overexpression of EVI1 and carries a dismal prognosis. Arsenic-containing compounds have been described to be efficacious in malignancies overexpressing EVI1. Here, we describe a case of AML with inv(3)(q21q26.2) treated with the organic arsenical darinaparsin. Using a "personalized medicine approach," two different arsenicals were screened for anti-leukemic effect against the patient's cells ex vivo. The most promising compound, darinaparsin, was selected for in vivo treatment. Clinical effect was almost immediate, with a normalization of temperature, a stabilization of white blood cell (WBC) counts and an increased quality of life. Longitudinal monitoring of patient response and resistance incorporating significant correlative studies on patient-derived blood samples over the two cycles of darinaparsin given to this patient allowed us to evaluate potential mechanisms of response and resistance. The anti-leukemic effects of darinaparsin correlated with inhibition of the alternative NF-κB pathway and production of the inflammatory cytokine IL-8. Emergence of resistance was suspected during treatment cycle 2 and supported by xenograft studies in nude mice. Darinaparsin resistance correlated with an attenuation of the effect of treatment on the alternative NF-κB pathway. The results from this patient indicate that darinaparsin may be a good treatment option for inv(3) AML and that inhibition of the alternative NF-κB pathway may be predictive of response. Longitudinal monitoring of disease response as well as several correlative parameters allowed for the generation of novel correlations and predictors of response to experimental therapy in a heavily pretreated patient.

7.
Cancers (Basel) ; 4(4): 1161-79, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-24213503

ABSTRACT

Acute myeloid leukemia (AML) is nearly always a fatal malignancy. For the past 40 years, the standard of care remains a combination of cytarabine and an anthracycline known as 7 + 3. This treatment regimen is troubled by both low survival rates (10% at 5 years) and deaths due to toxicity. Substantial new laboratory findings over the past decade have identified many cellular pathways that contribute to leukemogenesis. These studies have led to the development of novel agents designed to target these pathways. Here we discuss the molecular underpinnings and clinical benefits of these novel treatment strategies. Most importantly these studies demonstrate that clinical response is best achieved by stratifying each patient based on a detailed understanding of their molecular abnormalities.

8.
Front Pharmacol ; 2: 59, 2011.
Article in English | MEDLINE | ID: mdl-22007174

ABSTRACT

Therapeutic resistance remains a major cause of cancer-related deaths. Resistance can occur from the outset of treatment or as an acquired phenomenon after an initial clinical response. Therapeutic resistance is an almost universal phenomenon in the treatment of metastatic cancers. The advent of molecularly targeted treatments brought greater efficacy in patients whose tumors express a particular target or molecular signature. However, resistance remains a predictable challenge. This article provides an overview of somatic genomic events that confer resistance to cancer therapies. Some examples, including BCR-Abl, EML4-ALK, and the androgen receptor, contain mutations in the target itself, which hamper binding and inhibitory functions of therapeutic agents. There are also examples of somatic genetic changes in other genes or pathways that result in resistance by circumventing the inhibitor, as in resistance to trastuzumab and BRAF inhibitors. Yet other examples results in activation of cytoprotective genes. The fact that all of these mechanisms of resistance are due to somatic changes in the tumor's genome makes targeting them selectively a feasible goal. To identify and validate these changes, it is important to obtain biopsies of clinically resistant tumors. A rational consequence of this evolving knowledge is the growing appreciation that combinations of inhibitors will be needed to anticipate and overcome therapeutic resistance.

9.
Clin Cancer Res ; 17(9): 2874-84, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21415224

ABSTRACT

PURPOSE: We have evaluated the eukaryotic translation initiation factor 4E (eIF4E) as a potential biomarker and therapeutic target in breast cancer. eIF4E facilitates nuclear export and translation of specific, growth-stimulatory mRNAs and is frequently overexpressed in cancer. EXPERIMENTAL DESIGN: Breast cancer cells were treated with ribavirin, an inhibitor of eIF4E, and effects on cell proliferation and on known mRNA targets of eIF4E were determined. eIF4E expression was assessed, at the mRNA and protein level, in breast cancer cell lines and in skin biopsies from patients with metastatic disease. Additionally, pooled microarray data from 621 adjuvant untreated, node-negative breast cancers were analyzed for eIF4E expression levels and correlation with distant metastasis-free survival (DMFS), overall and within each intrinsic breast cancer subtype. RESULTS: At clinically relevant concentrations, ribavirin reduced cell proliferation and suppressed clonogenic potential, correlating with reduced mRNA export and protein expression of important eIF4E targets. This effect was suppressed by knockdown of eIF4E. Although eIF4E expression is elevated in all breast cancer cell lines, variability in ribavirin responsiveness was observed, indicating that other factors contribute to an eIF4E-dependent phenotype. Assessment of the prognostic value of high eIF4E mRNA in patient tumors found that significant discrimination between good and poor outcome groups was observed only in luminal B cases, suggesting that a specific molecular profile may predict response to eIF4E-targeted therapy. CONCLUSIONS: Inhibition of eIF4E is a potential breast cancer therapeutic strategy that may be especially promising against specific molecular subtypes and in metastatic as well as primary tumors.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Carcinoma/diagnosis , Carcinoma/drug therapy , Eukaryotic Initiation Factor-4E/genetics , Ribavirin/therapeutic use , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/classification , Breast Neoplasms/genetics , Carcinoma/classification , Carcinoma/genetics , Cell Line, Tumor , Cells, Cultured , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4E/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Organ Specificity/genetics , Prognosis , RNA, Small Interfering/pharmacology , Ribavirin/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
10.
Blood ; 114(2): 257-60, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19433856

ABSTRACT

The eukaryotic translation initiation factor eIF4E is elevated in 30% of malignancies including M4/M5 subtypes of acute myeloid leukemia (AML). The oncogenic potential of eIF4E arises from its ability to bind the 7-methyl guanosine (m(7)G) cap on mRNAs, thereby selectively enhancing eIF4E-dependent nuclear mRNA export and translation. We tested the clinical efficacy of targeting eIF4E in M4/M5 AML patients with a physical mimic of the m(7)G cap, ribavirin. Among 11 evaluable patients there were 1 complete remission (CR), 2 partial remissions (PRs), 2 blast responses (BRs), 4 stable diseases (SDs), and 2 progressive diseases (PDs). Ribavirin-induced relocalization of nuclear eIF4E to the cytoplasm and reduction of eIF4E levels were associated with clinical response. Lack of response or relapse coincided with continued or renewed nuclear localization of eIF4E. This first clinical study to target eIF4E in human malignancy demonstrates clinical activity and associated molecular responses in leukemic blasts. This trial is registered at ClinicalTrials.gov (NCT00559091).


Subject(s)
Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Ribavirin/therapeutic use , Adult , Aged , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged
11.
Mol Cell Biol ; 29(10): 2505-20, 2009 May.
Article in English | MEDLINE | ID: mdl-19273609

ABSTRACT

Characterizing mechanisms regulating mammary cell growth and differentiation is vital, as they may contribute to breast carcinogenesis. Here, we examine a cross talk mechanism(s) downstream of prolactin (PRL), a primary differentiation hormone, and epidermal growth factor (EGF), an important proliferative factor, in mammary epithelial cell growth and differentiation. Our data indicate that EGF exerts inhibitory effects on PRL-induced cellular differentiation by interfering with Stat5a-mediated gene expression independent of the PRL-proximal signaling cascade. Additionally, our data show that PRL is a potent inhibitor of EGF-induced cell proliferation. We identify tyrosine phosphorylation of the growth factor receptor-bound protein 2 (Grb2) as a critical mechanism by which PRL antagonizes EGF-induced cell proliferation by attenuating the activation of the Ras/mitogen-activated protein kinase (MAPK) pathway. Together, our results define a novel negative cross-regulation between PRL and EGF involving the Jak2/Stat5a and Ras/MAPK pathways through tyrosine phosphorylation of Grb2.


Subject(s)
Cell Differentiation/physiology , Epidermal Growth Factor/metabolism , Epithelial Cells/physiology , GRB2 Adaptor Protein/metabolism , MAP Kinase Signaling System/physiology , Mammary Glands, Human/cytology , Prolactin/metabolism , Animals , Breast Neoplasms/metabolism , Cell Communication/physiology , Cell Line , Cell Proliferation , Enzyme Activation , Epithelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases , Female , GRB2 Adaptor Protein/genetics , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mammary Glands, Human/physiology , Phosphorylation , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tyrosine/metabolism , ras Proteins/genetics , ras Proteins/metabolism
12.
J Biol Chem ; 283(3): 1293-1307, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18024957

ABSTRACT

Both the transforming growth factor-beta (TGFbeta)/Smad and the prolactin/JAK/STAT pathway are critical to the proper development, maintenance, and function of the mammary epithelial tissue. Interestingly, opposing physiological effects between these two signaling pathways are prominent in the regulation of mammary gland development. However, the exact nature of the biological network existing between the Smad and STAT signal transduction pathways has remained elusive. We identified a novel regulatory cross-talk mechanism by which TGFbeta-induced Smad signaling acts to antagonize prolactin-mediated JAK/STAT signaling and expression of target genes. Furthermore, we found activin, another member of the TGFbeta family, to also efficiently block STAT5 signaling and beta-casein expression in mammary epithelial cells. Our results indicate that ligand-induced activation of Smad2, -3, and -4 by activin and TGFbeta leads to a direct inhibition of STAT5 transactivation and STAT5-mediated transcription of the downstream target genes, beta-casein and cyclin D1, thereby blocking vital processes for mammary gland growth and differentiation. Finally, we unveiled the mechanism by which these two signaling cascades antagonize their effects, and we found that activated Smads inhibit STAT5 association with its co-activator CREB-binding protein, thus blocking STAT5 transactivation of its target genes and leading to inhibition of mammary gland differentiation and lactation.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Mammary Glands, Animal/cytology , STAT5 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Transcription, Genetic , Activins/metabolism , Animals , Caseins/metabolism , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin D1/genetics , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Female , Gene Expression Regulation/drug effects , Humans , Janus Kinases/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/enzymology , Milk Proteins/genetics , Milk Proteins/metabolism , Phosphorylation/drug effects , Prolactin/pharmacology , Promoter Regions, Genetic , Protein Binding/drug effects , Protein Transport/drug effects , Repressor Proteins/metabolism , Signal Transduction/drug effects , Thermodynamics , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , p300-CBP Transcription Factors/metabolism
13.
Cancer Res ; 66(3): 1824-32, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452244

ABSTRACT

Prolactin hormone (PRL) is well characterized as a terminal differentiation factor for mammary epithelial cells and as an autocrine growth/survival factor in breast cancer cells. However, this function of PRL may not fully signify its role in breast tumorigenesis. Cancer is a complex multistep progressive disease resulting not only from defects in cell growth but also in cell differentiation. Indeed, dedifferentiation of tumor cells is now recognized as a crucial event in invasion and metastasis. PRL plays a critical role in inducing/maintaining differentiation of mammary epithelial cells, suggesting that PRL signaling could serve to inhibit tumor progression. We show here that in breast cancer cells, PRL and Janus-activated kinase 2, a major kinase involved in PRL signaling, play a critical role in regulating epithelial-mesenchymal transformation (EMT), an essential process associated with tumor metastasis. Activation of the PRL receptor (PRLR), achieved by restoring PRL/JAK2 signaling in mesenchymal-like breast cancer cells, MDA-MB-231, suppressed their mesenchymal properties and reduced their invasive behavior. While blocking PRL autocrine function in epithelial-like breast cancer cells, T47D, using pharmacologic and genetic approaches induced mesenchymal-like phenotypic changes and enhanced their invasive propensity. Moreover, our results indicate that blocking PRL signaling led to activation of mitogen-activated protein kinase (extracellular signal-regulated kinase 1/2) and transforming growth factor-beta/Smad signaling pathways, two major prometastatic pathways. Furthermore, our results indicate that following PRL/JAK2 inhibition, ERK1/2 activation precedes and is required for Smad2 activation and EMT induction in breast cancer cells. Together, these results highlight PRL as a critical regulator of epithelial plasticity and implicate PRL as an invasion suppressor hormone in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Prolactin/physiology , Animals , Breast Neoplasms/enzymology , CHO Cells , Cricetinae , Enzyme Activation , Epithelial Cells/pathology , Humans , Janus Kinase 2 , MAP Kinase Signaling System/physiology , Mesoderm/pathology , Neoplasm Invasiveness , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Smad Proteins/metabolism
14.
EMBO J ; 24(18): 3247-58, 2005 Sep 21.
Article in English | MEDLINE | ID: mdl-16121194

ABSTRACT

Signaling from the activin/transforming growth factor beta (TGFbeta) family of cytokines is a tightly regulated process. Disregulation of TGFbeta signaling is often the underlying basis for various cancers, tumor metastasis, inflammatory and autoimmune diseases. In this study, we identify the protein G-coupled receptor kinase 2 (GRK2), a kinase involved in the desensitization of G protein-coupled receptors (GPCR), as a downstream target and regulator of the TGFbeta-signaling cascade. TGFbeta-induced expression of GRK2 acts in a negative feedback loop to control TGFbeta biological responses. Upon TGFbeta stimulation, GRK2 associates with the receptor-regulated Smads (R-Smads) through their MH1 and MH2 domains and phosphorylates their linker region. GRK2 phosphorylation of the R-Smads inhibits their carboxyl-terminal, activating phosphorylation by the type I receptor kinase, thus preventing nuclear translocation of the Smad complex, leading to the inhibition of TGFbeta-mediated target gene expression, cell growth inhibition and apoptosis. Furthermore, we demonstrate that GRK2 antagonizes TGFbeta-induced target gene expression and apoptosis ex vivo in primary hepatocytes, establishing a new role for GRK2 in modulating single-transmembrane serine/threonine kinase receptor-mediated signal transduction.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Active Transport, Cell Nucleus , Activins/pharmacology , Animals , Apoptosis/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , G-Protein-Coupled Receptor Kinase 2 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Norepinephrine/pharmacology , Phosphorylation , Protein Binding , Rats , Smad2 Protein , Smad3 Protein , Substrate Specificity , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , beta-Adrenergic Receptor Kinases
15.
Nat Cell Biol ; 4(12): 963-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12447389

ABSTRACT

Members of the transforming growth factor beta (TGF-beta) family regulate fundamental physiological processes, such as cell growth, differentiation and apoptosis, in almost all cell types. As a result, defects in TGF-beta signalling pathways have been linked to uncontrolled cellular proliferation and carcinogenesis. Here, we explored the signal transduction mechanisms downstream of the activin/TGF-beta receptors that result in cell growth arrest and apoptosis. We show that in haematopoietic cells, TGF-beta family members regulate apoptosis through expression of the inositol phosphatase SHIP (Src homology 2 (SH2) domain-containing 5' inositol phosphatase), a central regulator of phospholipid metabolism. We also demonstrated that the Smad pathway is required in the transcriptional regulation of the SHIP gene. Activin/TGF-beta-induced expression of SHIP results in intracellular changes in the pool of phospholipids, as well as in inhibition of both Akt/PKB (protein kinase B) phosphorylation and cell survival. Our results link phospholipid metabolism to activin/TGF-beta-mediated apoptosis and define TGF-beta family members as potent inducers of SHIP expression.


Subject(s)
Activins/physiology , Apoptosis/physiology , Phosphoric Monoester Hydrolases/biosynthesis , Transforming Growth Factor beta/physiology , Animals , Hematopoiesis/physiology , Humans , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/physiology , Signal Transduction/physiology , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...