18 results on '"Mark A. Titus"'
Search Results
2. Data from The Efflux Transporter ABCG2 Maintains Prostate Stem Cells
- Author
-
Wendy J. Huss, Mark A. Titus, Austin Miller, and Neha G. Sabnis
- Abstract
Prostate stem cells (PSC) are characterized by their intrinsic resistance to androgen deprivation therapy (ADT), possibly due to the lack of androgen receptor (AR) expression. PSCs resistance to ADT and PSC expansion in castration resistant prostate cancer (CRPC) has sparked great interest in using differentiation therapy as an adjuvant to ADT. Understanding the mechanisms, by which PSCs maintain their undifferentiated phenotype, thus has important implications in differentiation therapy. In the prostate, the ATP binding cassette sub-family G member 2 (ABCG2) transporters, which enrich for AR-positive, ADT-resistant PSCs, play an important role in regulating the intracellular androgen levels by effluxing androgens. We hypothesized that the ABCG2-mediated androgen efflux is responsible for maintaining PSCs in an undifferentiated state. Using the HPr-1-AR (nontumorigenic) and CWR-R1 (tumorigenic) prostate cell lines, it was demonstrated that inhibiting the ABCG2-mediated androgen efflux, with Ko143 (ABCG2 inhibitor), increased the nuclear AR expression due to elevated intracellular androgen levels. Increased nuclear translocation of AR is followed by increased expression of AR regulated genes, a delayed cell growth response, and increased luminal differentiation. Furthermore, Ko143 reduced tumor growth rates in mice implanted with ABCG2-expressing CWR-R1 cells. In addition, Ko143-treated mice had more differentiated tumors as evidenced by an increased percentage of CK8+/AR+ luminal cells and decreased percentage of ABCG2-expressing cells. Thus, inhibiting ABCG2-mediated androgen efflux forces the PSCs to undergo an AR-modulated differentiation to an ADT-sensitive luminal phenotype.Implications: This study identifies the mechanism by which the prostate stem cell marker, ABCG2, plays a role in prostate stem cell maintenance and provides a rationale for targeting ABCG2 for differentiation therapy in prostate cancer. Mol Cancer Res; 15(2); 128–40. ©2016 AACR.
- Published
- 2023
3. Supplementary Figure Legend from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Supplementary Figure Legend
- Published
- 2023
4. Data from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer. AR expression has also been reported in other solid tumors, including renal cell carcinoma (RCC), but its biological role here remains unclear. Through integrative analysis of a reverse phase protein array, we discovered increased expression of AR in an RCC patient–derived xenograft model of acquired resistance to the receptor tyrosine kinase inhibitor (RTKi) sunitinib. AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro. An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Enzalutamide-induced AR degradation was rescued by either proteasome inhibition or by knockdown of the AR ubiquitin ligase speckle-type POZ protein (SPOP). In vivo treatment with enzalutamide and sunitinib demonstrated that this combination efficiently induced tumor regression in a RCC model following acquired sunitinib resistance. Overall, our results suggest the potential role of AR as a target for therapeutic interventions, in combination with RTKi, to overcome drug resistance in RCC.Significance: These findings highlight the therapeutic potential of targeting the androgen receptor to overcome RCC resistance to receptor tyrosine kinase inhibitors. Cancer Res; 78(11); 2886–96. ©2018 AACR.
- Published
- 2023
5. Data from A Phase II Study of Cabozantinib and Androgen Ablation in Patients with Hormone-Naïve Metastatic Prostate Cancer
- Author
-
Christopher Logothetis, Patricia Troncoso, Eleni Efstathiou, Gary E. Gallick, Theocharis Panaretakis, Mark A. Titus, Sumankalai Ramachandran, Guocan Wang, Sue-Hwa Lin, Nora M. Navone, Shixia Huang, Kimal Rajapakshe, Cristian Coarfa, Ana M. Aparicio, Shi-Ming Tu, Sumit K. Subudhi, Amado J. Zurita, Lianchun Xiao, Graciela M. Nogueras-Gonzalez, Miao Zhang, and Paul G. Corn
- Abstract
Purpose:Cabozantinib, an oral inhibitor of c-MET/VEGFR2 signaling, improved progression-free survival (mPFS) but not overall survival (OS) in metastatic castrate-resistant prostate cancer. We evaluated cabozantinib plus androgen deprivation therapy (ADT) in hormone-naïve metastatic prostate cancer (HNMPCa).Patients and Methods:Patients received ADT plus cabozantinib starting at 60 mg daily. The primary endpoint was castrate-resistant PFS by radiographic criteria, clinical progression, or receipt of additional therapy. Secondary endpoints included OS, safety, radiographic responses, and biomarker modulation.Results:Sixty-two patients received treatment. With a median follow-up of 31.2 months, the mPFS was 16.1 months (95% CI, 14.6–22.7 months), and mOS was not reached. Reductions in PSA ≥ 90%, bone-specific alkaline phosphatase ≥ 50%, and urine N-telopeptides ≥ 50% occurred in 83%, 87%, and 86% of evaluable patients, respectively. Responses in bone scan and measurable disease were observed in 81% of and 90% of evaluable patients, respectively. Most common grade 3 adverse events were hypertension (19%), diarrhea (6%), and thromboembolic events (6%), and dose reductions occurred in 85% of patients. Analysis of baseline cytokine and angiogenic factors (CAFs) revealed that higher plasma concentrations of Lumican, CXCL5, CD25, and CD30 were associated with shorter PFS as was high tumor expression of pFGFR1.Conclusions:Cabozantinib plus ADT has promising clinical activity in HNMPCa. CAF profiles and tissue markers suggest candidate prognostic and predictive markers of cabozantinib benefit and provide insights for rational therapy combinations.
- Published
- 2023
6. Supplementary Video S2. from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Enzalutamide prevents Sunitinib induced AR nuclear localization. Real-time fluorescence live cell imaging of 786-0 cells expressing AR-EGFP pretreated with 500 nM Enzalutamide for 30 minutes does not show increase in AR nuclear localization following Sunitinib treatment (5uM Sunitinib t=30s).
- Published
- 2023
7. Data from Organelle-Derived Acetyl-CoA Promotes Prostate Cancer Cell Survival, Migration, and Metastasis via Activation of Calmodulin Kinase II
- Author
-
Sue-Hwa Lin, Leta K. Nutt, Mark A. Titus, Gary E. Gallick, Li-Yuan Yu-Lee, Daniel E. Frigo, Yu-Chen Lee, Song-Chang Lin, Chien-Jui Cheng, and Guoyu Yu
- Abstract
Although emerging evidence suggests a potential role of calcium/calmodulin-dependent kinase II (CaMKII) in prostate cancer, its role in prostate cancer tumorigenesis is largely unknown. Here, we examine whether the acetyl CoA-CaMKII pathway, first described in frog oocytes, promotes prostate cancer tumorigenesis. In human prostate cancer specimens, metastatic prostate cancer expressed higher levels of active CaMKII compared with localized prostate cancer. Correspondingly, basal CaMKII activity was significantly higher in the more tumorigenic PC3 and PC3-mm2 cells relative to the less tumorigenic LNCaP and C4-2B4 cells. Deletion of CaMKII by CRISPR/Cas9 in PC3-mm2 cells abrogated cell survival under low-serum conditions, anchorage-independent growth and cell migration; overexpression of constitutively active CaMKII in C4-2B4 cells promoted these phenotypes. In an animal model of prostate cancer metastasis, genetic ablation of CaMKII reduced PC3-mm2 cell metastasis from the prostate to the lymph nodes. Knockdown of the acetyl-CoA transporter carnitine acetyltransferase abolished CaMKII activation, providing evidence that acetyl-CoA generated from organelles is a major activator of CaMKII. Genetic deletion of the β-oxidation rate-limiting enzyme ACOX family proteins decreased CaMKII activation, whereas overexpression of ACOXI increased CaMKII activation. Overall, our studies identify active CaMKII as a novel connection between organelle β-oxidation and acetyl-CoA transport with cell survival, migration, and prostate cancer metastasis.Significance: This study identifies a cell metabolic pathway that promotes prostate cancer metastasis and suggests prostate cancer may be susceptible to β-oxidation inhibitors. Cancer Res; 78(10); 2490–502. ©2018 AACR.
- Published
- 2023
8. Supplementary Data from Organelle-Derived Acetyl-CoA Promotes Prostate Cancer Cell Survival, Migration, and Metastasis via Activation of Calmodulin Kinase II
- Author
-
Sue-Hwa Lin, Leta K. Nutt, Mark A. Titus, Gary E. Gallick, Li-Yuan Yu-Lee, Daniel E. Frigo, Yu-Chen Lee, Song-Chang Lin, Chien-Jui Cheng, and Guoyu Yu
- Abstract
This file contains immunofluorescence of LNCaP, C4-2B4, PC3, and PC3-mm2 PCa cell lines (Supplementary Fig.S1); Immunohistochemistry for the expression of pCaMKII (T286) in specimens from human primary prostate tumor, lymph node metastasis, and bone metastasis (Supplementary Fig. S2); Re-expression of constitutively active CaMKII in PC3-mm2 cells with knockout of CaMKII (Supplementary Fig. S3); Effect of re-expression of constitutively active CaMKII in PC3-mm2 cells with knockout of CaMKII on prostate cancer metastasis (Supplementary Fig. S4); Knockdown of AcoxI in PC3-mm2 cells (Supplementary Fig. S5); Effect of AcoxI-III knockout on the levels of individuallipid components (Supplementary Fig. S6) and Sequence of primers and gDNA of CaMKII and ACOX (Table S1).
- Published
- 2023
9. Supplementary Data from A Phase II Study of Cabozantinib and Androgen Ablation in Patients with Hormone-Naïve Metastatic Prostate Cancer
- Author
-
Christopher Logothetis, Patricia Troncoso, Eleni Efstathiou, Gary E. Gallick, Theocharis Panaretakis, Mark A. Titus, Sumankalai Ramachandran, Guocan Wang, Sue-Hwa Lin, Nora M. Navone, Shixia Huang, Kimal Rajapakshe, Cristian Coarfa, Ana M. Aparicio, Shi-Ming Tu, Sumit K. Subudhi, Amado J. Zurita, Lianchun Xiao, Graciela M. Nogueras-Gonzalez, Miao Zhang, and Paul G. Corn
- Abstract
Additional Figures and Tables
- Published
- 2023
10. Data from 14-3-3η Amplifies Androgen Receptor Actions in Prostate Cancer
- Author
-
Frank S. French, James L. Mohler, Elizabeth M. Wilson, Haian Fu, Romesh R. Subramanian, O. Harris Ford, Christopher W. Gregory, Jiann-an Tan, and Mark A. Titus
- Abstract
Purpose: Androgen receptor abundance and androgen receptor–regulated gene expression in castration-recurrent prostate cancer are indicative of androgen receptor activation in the absence of testicular androgen. Androgen receptor transactivation of target genes in castration-recurrent prostate cancer occurs in part through mitogen signaling that amplifies the actions of androgen receptor and its coregulators. Herein we report on the role of 14-3-3η in androgen receptor action.Experimental Design and Results: Androgen receptor and 14-3-3η colocalized in COS cell nuclei with and without androgen, and 14-3-3η promoted androgen receptor nuclear localization in the absence of androgen. 14-3-3η interacted with androgen receptor in cell-free binding and coimmunoprecipitation assays. In the recurrent human prostate cancer cell line, CWR-R1, native endogenous androgen receptor transcriptional activation was stimulated by 14-3-3η at low dihydrotestosterone concentrations and was increased by epidermal growth factor. Moreover, the dihydrotestosterone- and epidermal growth factor–dependent increase in androgen receptor transactivation was inhibited by a dominant negative 14-3-3η. In the CWR22 prostate cancer xenograft model, 14-3-3η expression was increased by androgen, suggesting a feed-forward mechanism that potentiates both 14-3-3η and androgen receptor actions. 14-3-3η mRNA and protein decreased following castration of tumor-bearing mice and increased in tumors of castrate mice after treatment with testosterone. CWR22 tumors that recurred 5 months after castration contained 14-3-3η levels similar to the androgen-stimulated tumors removed before castration. In a human prostate tissue microarray of clinical specimens, 14-3-3η localized with androgen receptor in nuclei, and the similar amounts expressed in castration-recurrent prostate cancer, androgen-stimulated prostate cancer, and benign prostatic hyperplasia were consistent with androgen receptor activation in recurrent prostate cancer.Conclusion: 14-3-3η enhances androgen- and mitogen-induced androgen receptor transcriptional activity in castration-recurrent prostate cancer. (Clin Cancer Res 2009;15(24):7571–81)
- Published
- 2023
11. Supplementary Figures from Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma
- Author
-
Roberto Pili, Chinghai Kao, Mark A. Titus, Sreenivasulu Chintala, Sreevani Arisa, Ashley R. Orillion, Nur P. Damayanti, and Remi Adelaiye-Ogala
- Abstract
Enzalutamide modulates the protein expression of sunitinib-induced AR regulated genes.
- Published
- 2023
12. Ablating Lgr5-expressing prostatic stromal cells activates the ERK-mediated mechanosensory signaling and disrupts prostate tissue homeostasis
- Author
-
Xing Wei, Li Zhang, Yiqun Zhang, Cody Cooper, Chris Brewer, Chia-Feng Tsai, Yi-Ting Wang, Micah Glaz, Hunter B. Wessells, Jianwen Que, Mark A. Titus, Vincenzino Cirulli, Adam Glaser, Tao Liu, Nicholas P. Reder, Chad J. Creighton, and Li Xin
- Subjects
Male ,Mice ,Prostate ,Animals ,Homeostasis ,Stromal Cells ,General Biochemistry, Genetics and Molecular Biology ,Receptors, G-Protein-Coupled ,Signal Transduction - Abstract
Functional implication of stromal heterogeneity in the prostate remains incompletely understood. Using lineage tracing and light-sheet imaging, we show that some fibroblast cells at the mouse proximal prostatic ducts and prostatic urethra highly express Lgr5. Genetic ablation of these anatomically restricted stromal cells, but not nonselective ablation of prostatic stromal cells, rapidly induces prostate epithelial turnover and dedifferentiation that are reversed following spontaneous restoration of the Lgr5
- Published
- 2022
13. The prolyl isomerase pin1 regulates mRNA levels of genes with short half-lives by targeting specific RNA binding proteins.
- Author
-
Nithya Krishnan, Mark A Titus, and Roopa Thapar
- Subjects
Medicine ,Science - Abstract
The peptidyl-prolyl isomerase Pin1 is over-expressed in several cancer tissues is a potential prognostic marker in prostate cancer, and Pin1 ablation can suppress tumorigenesis in breast and prostate cancers. Pin1 can co-operate with activated ErbB2 or Ras to enhance tumorigenesis. It does so by regulating the activity of proteins that are essential for gene expression and cell proliferation. Several targets of Pin1 such as c-Myc, the Androgen Receptor, Estrogen Receptor-alpha, Cyclin D1, Cyclin E, p53, RAF kinase and NCOA3 are deregulated in cancer. At the posttranscriptional level, emerging evidence indicates that Pin1 also regulates mRNA decay of histone mRNAs, GM-CSF, Pth, and TGFβ mRNAs by interacting with the histone mRNA specific protein SLBP, and the ARE-binding proteins AUF1 and KSRP, respectively. To understand how Pin1 may affect mRNA abundance on a genome-wide scale in mammalian cells, we used RNAi along with DNA microarrays to identify genes whose abundance is significantly altered in response to a Pin1 knockdown. Functional scoring of differentially expressed genes showed that Pin1 gene targets control cell adhesion, leukocyte migration, the phosphatidylinositol signaling system and DNA replication. Several mRNAs whose abundance was significantly altered by Pin1 knockdown contained AU-rich element (ARE) sequences in their 3' untranslated regions. We identified HuR and AUF1 as Pin1 interacting ARE-binding proteins in vivo. Pin1 was also found to stabilize all core histone mRNAs in this study, thereby validating our results from a previously published study. Statistical analysis suggests that Pin1 may target the decay of essential mRNAs that are inherently unstable and have short to medium half-lives. Thus, this study shows that an important biological role of Pin1 is to regulate mRNA abundance and stability by interacting with specific RNA-binding proteins that may play a role in cancer progression.
- Published
- 2014
- Full Text
- View/download PDF
14. Dominant-negative androgen receptor inhibition of intracrine androgen-dependent growth of castration-recurrent prostate cancer.
- Author
-
Mark A Titus, Brian Zeithaml, Boris Kantor, Xiangping Li, Karin Haack, Dominic T Moore, Elizabeth M Wilson, James L Mohler, and Tal Kafri
- Subjects
Medicine ,Science - Abstract
Prostate cancer (CaP) is the second leading cause of cancer death in American men. Androgen deprivation therapy is initially effective in CaP treatment, but CaP recurs despite castrate levels of circulating androgen. Continued expression of the androgen receptor (AR) and its ligands has been linked to castration-recurrent CaP growth.In this report, the ligand-dependent dominant-negative ARΔ142-337 (ARΔTR) was expressed in castration-recurrent CWR-R1 cell and tumor models to elucidate the role of AR signaling. Expression of ARΔTR decreased CWR-R1 tumor growth in the presence and absence of exogenous testosterone (T) and improved survival in the presence of exogenous T. There was evidence for negative selection of ARΔTR transgene in T-treated mice. Mass spectrometry revealed castration-recurrent CaP dihydrotestosterone (DHT) levels sufficient to activate AR and ARΔTR. In the absence of exogenous testosterone, CWR-R1-ARΔTR and control cells exhibited altered androgen profiles that implicated epithelial CaP cells as a source of intratumoral AR ligands.The study provides in vivo evidence that activation of AR signaling by intratumoral AR ligands is required for castration-recurrent CaP growth and that epithelial CaP cells produce sufficient active androgens for CaP recurrence during androgen deprivation therapy. Targeting intracrine T and DHT synthesis should provide a mechanism to inhibit AR and growth of castration-recurrent CaP.
- Published
- 2012
- Full Text
- View/download PDF
15. 5α-reductase type 3 enzyme in benign and malignant prostate
- Author
-
Mark A, Titus, Yun, Li, Olga G, Kozyreva, Varun, Maher, Alejandro, Godoy, Gary J, Smith, and James L, Mohler
- Subjects
Male ,Cholestenone 5 alpha-Reductase ,Prostate ,Prostatic Hyperplasia ,Gene Expression ,Prostatic Neoplasms ,CHO Cells ,Dutasteride ,Recombinant Proteins ,Article ,Isoenzymes ,Mice ,Prostatic Neoplasms, Castration-Resistant ,Cricetulus ,3-Oxo-5-alpha-Steroid 4-Dehydrogenase ,Azasteroids ,Androgens ,Animals ,Heterografts ,Humans ,RNA, Messenger ,Enzyme Inhibitors ,Neoplasm Transplantation - Abstract
Currently available 5α-reductase inhibitors are not completely effective for treatment of benign prostate enlargement, prevention of prostate cancer (CaP), or treatment of advanced castration-recurrent (CR) CaP. We tested the hypothesis that a novel 5α-reductase, 5α-reductase-3, contributes to residual androgen metabolism, especially in CR-CaP.A new protein with potential 5α-reducing activity was expressed in CHO-K1 cellsandTOP10 E. coli for characterization. Protein lysates and total mRNA were isolated from preclinical and clinical tissues. Androgen metabolism was assessed using androgen precursors and thin layer chromatography or liquid chromatography tandem mass spectrometry.The relative mRNA expression for the three 5α-reductase enzymes in clinical samples of CR-CaP was 5α-reductase-3 ≫ 5α-reductase-1 5α-reductase-2. Recombinant 5α-reductase-3 protein incubations converted testosterone, 4-androstene-3,17-dione (androstenedione) and 4-pregnene-3,20-dione (progesterone) to dihydrotestosterone, 5α-androstan-3,17-dione, and 5α-pregnan-3,20-dione, respectively. 5α-Reduced androgen metabolites were measurable in lysates from androgen-stimulated (AS) CWR22 and CR-CWR22 tumors and clinical specimens of AS-CaP and CR-CaP pre-incubated with dutasteride (a bi-specific inhibitor of 5α-reductase-1 and 2).Human prostate tissues contain a third 5α-reductase that was inhibited poorly by dutasteride at high androgen substrate concentration in vitro, and it may promote DHT formation in vivo, through alternative androgen metabolism pathways when testosterone levels are low.
- Published
- 2013
16. Leptin as a novel profibrogenic cytokine in hepatic stellate cells: mitogenesis and inhibition of apoptosis mediated by extracellular regulated kinase (Erk) and Akt phosphorylation
- Author
-
Jeffrey J. Floyd, Mark A. Titus, Shanthi Srinivasan, Frank A. Anania, Shanthi V. Sitaraman, Xiaokun Ding, and Neeraj K. Saxena
- Subjects
Leptin ,MAPK/ERK pathway ,Apoptosis ,Suppressor of Cytokine Signaling Proteins ,Biochemistry ,TNF-Related Apoptosis-Inducing Ligand ,chemistry.chemical_compound ,Cyclin D1 ,LY294002 ,SOCS3 ,Cycloheximide ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Cells, Cultured ,Membrane Glycoproteins ,Janus kinase 2 ,biology ,Caspase 3 ,digestive, oral, and skin physiology ,Cell biology ,Liver ,Caspases ,Receptors, Leptin ,hormones, hormone substitutes, and hormone antagonists ,Platelet-derived growth factor receptor ,Biotechnology ,DNA Replication ,Morpholines ,Mitosis ,Receptors, Cell Surface ,Protein Serine-Threonine Kinases ,Article ,Proto-Oncogene Proteins ,Genetics ,Molecular Biology ,Protein kinase B ,Cell Proliferation ,Flavonoids ,Leptin receptor ,Tumor Necrosis Factor-alpha ,DNA ,Repressor Proteins ,chemistry ,Chromones ,Suppressor of Cytokine Signaling 3 Protein ,biology.protein ,Cancer research ,Hepatic stellate cell ,Mitogens ,Apoptosis Regulatory Proteins ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
A key feature in the molecular pathogenesis of liver fibrosis requires maintenance of the activated hepatic stellate cell (HSC) phenotype by both proliferation and inhibition of apoptosis. We provide evidence that leptin is a potent HSC mitogen and dramatically inhibits stellate cell apoptosis. Leptin proved to be as potent an HSC mitogen as platelet-derived growth factor (PDGF) as assessed by bromodeoxyuridine (BrdU) incorporation in isolated primary HSCs; data using fluorescent propidium iodide (PI) uptake revealed that leptin, like PDGF, increased HSC populations in the S- and G2/M-phases of the cell cycle. Leptin resulted in a robust increase in cyclin D1 expression. Using the chemical inhibitor of Janus kinase 2 (Jak2) activity, AG 490, and overexpression of the suppressor of cytokine signaling 3 (SOCS-3), we show that blockade of leptin receptor (Ob-Rb) phosphorylation blocks leptin-induced HSC proliferation. Leptin-associated phosphorylation of both extracellular regulated kinase (p44/p42, Erk) and Akt is also prohibited. Further, the PI-3 kinase inhibitor LY294002 and MAPK inhibitor PD98059 were found to significantly reduce leptin-induced HSC proliferation, thereby indicating that leptin induced HSC proliferation is Akt- and Erk-dependent. Akt was also protective against HSC apoptosis. Leptin abolished both cycloheximide-induced and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, demonstrated by reduced caspase-3 activity, HSC-TUNEL staining, and DNA fragmentation. We conclude that leptin acts as a direct hepatic stellate cell survival agonist. Importantly, we have demonstrated that leptin-induced HSC proliferation and survival by Ob-Rb phosphorylation are both Erk- and Akt-dependent.
- Published
- 2004
17. [Untitled]
- Author
-
Xiaotao Duan, Jun Qu, Mark A. Titus, and James L. Mohler
- Subjects
Urology - Published
- 2011
18. Leptin signal transduction in hepatic stellate cells involves activation of the mitogen activated protein kinase (MAPK) p44/p42, or ERK, as well as the signal transducer and activator of transcription 3 (Stat 3)
- Author
-
Neeraj K. Saxena, Mark A. Titus, Frank A. Anania, and Jeffrey J. Floyd
- Subjects
MAPK/ERK pathway ,Hepatology ,biology ,Chemistry ,Leptin ,Gastroenterology ,stat ,Cell biology ,Mitogen-activated protein kinase ,biology.protein ,Hepatic stellate cell ,STAT protein ,Signal transduction ,STAT4 - Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.