Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Luowei Li is active.

Publication


Featured researches published by Luowei Li.


Molecular and Cellular Biology | 1999

Protein Kinase Cδ Targets Mitochondria, Alters Mitochondrial Membrane Potential, and Induces Apoptosis in Normal and Neoplastic Keratinocytes When Overexpressed by an Adenoviral Vector

Luowei Li; Patricia S. Lorenzo; Krisztina Bogi; Peter M. Blumberg; Stuart H. Yuspa

ABSTRACT Inactivation of protein kinase Cδ (PKCδ) is associated with resistance to terminal cell death in epidermal tumor cells, suggesting that activation of PKCδ in normal epidermis may be a component of a cell death pathway. To test this hypothesis, we constructed an adenovirus vector carrying an epitope-tagged PKCδ under a cytomegalovirus promoter to overexpress PKCδ in normal and neoplastic keratinocytes. While PKCδ overexpression was detected by immunoblotting in keratinocytes, the expression level of other PKC isozymes, including PKCα, PKCɛ, PKCζ, and PKCη, did not change. Calcium-independent PKC-specific kinase activity increased after infection of keratinocytes with the PKCδ adenovirus. Activation of PKCδ by 12-O-tetradecanoylphorbol-13-acetate (TPA) at a nanomolar concentration was lethal to normal and neoplastic mouse and human keratinocytes overexpressing PKCδ. Lethality was inhibited by PKC selective inhibitors, GF109203X and Ro-32-0432. TPA-induced cell death was apoptotic as evidenced by morphological criteria, TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) assay, DNA fragmentation, and increased caspase activity. Subcellular fractionation indicated that PKCδ translocated to a mitochondrial enriched fraction after TPA activation, and this finding was confirmed by confocal microscopy of cells expressing a transfected PKCδ-green fluorescent protein fusion protein. Furthermore, activation of PKCδ in keratinocytes altered mitochondrial membrane potential, as indicated by rhodamine-123 fluorescence. Mitochondrial inhibitors, rotenone and antimycin A, reduced TPA-induced cell death in PKCδ-overexpressing keratinocytes. These results indicate that PKCδ can initiate a death pathway in keratinocytes that involves direct interaction with mitochondria and alterations of mitochondrial function.


Journal of Biological Chemistry | 2000

Involvement of Protein Kinase C δ (PKCδ) in Phorbol Ester-induced Apoptosis in LNCaP Prostate Cancer Cells LACK OF PROTEOLYTIC CLEAVAGE OF PKCδ

Teruhiko Fujii; Maria Laura Garcia-Bermejo; Juan Lucas Bernabó; Jorge Caamano; Motoi Ohba; Toshio Kuroki; Luowei Li; Stuart H. Yuspa; Marcelo G. Kazanietz

Phorbol esters, the activators of protein kinase C (PKC), induce apoptosis in androgen-sensitive LNCaP prostate cancer cells. The role of individual PKC isozymes as mediators of this effect has not been thoroughly examined to date. To study the involvement of the novel isozyme PKCδ, we used a replication-deficient adenovirus (PKCδAdV), which allowed for a tightly controlled expression of PKCδ in LNCaP cells. A significant reduction in cell number was observed after infection of LNCaP cells with PKCδAdV. Overexpression of PKCδ markedly enhanced the apoptotic effect of phorbol 12-myristate 13-acetate in LNCaP cells. PKCδ-mediated apoptosis was substantially reduced by the pan-caspase inhibitor z-VAD and by Bcl-2 overexpression. Importantly, and contrary to other cell types, PKCδ-mediated apoptosis does not involve its proteolytic cleavage by caspase-3, suggesting that allosteric activation of PKCδ is sufficient to trigger apoptosis in LNCaP cells. In addition, phorbol ester-induced apoptosis was blocked by a kinase-deficient mutant of PKCδ, supporting the concept that PKCδ plays an important role in the regulation of apoptotic cell death in LNCaP prostate cancer cells.


Journal of Biological Chemistry | 2006

Protein Kinase C Negatively Regulates Akt Activity and Modifies UVC-induced Apoptosis in Mouse Keratinocytes

Luowei Li; Keeran Sampat; Nancy Hu; Julia Zakari; Stuart H. Yuspa

Skin keratinocytes are subject to frequent chemical and physical injury and have developed elaborate cell survival mechanisms to compensate. Among these, the Akt/protein kinase B (PKB) pathway protects keratinocytes from the toxic effects of ultraviolet light (UV). In contrast, the protein kinase C (PKC) family is involved in several keratinocyte death pathways. During an examination of potential interactions among these two pathways, we found that the insulin-like growth factor (IGF-1) activates both the PKC and the Akt signaling pathways in cultured primary mouse keratinocytes as indicated by increased phospho-PKC and phospho-Ser-473-Akt. IGF-1 also selectively induced translocation of PKCδ and PKCϵ from soluble to particulate fractions in mouse keratinocytes. Furthermore, the PKC-specific inhibitor, GF109203X, increased IGF-1-induced phospho-Ser-473-Akt and Akt kinase activity and enhanced IGF-1 protection from UVC-induced apoptosis. Selective activation of PKC by 12-O-tetradecanoylphorbol-13-acetate (TPA) reduced phospho-Ser-473-Akt, suggesting that activation of PKC inhibits Akt activity. TPA also attenuated IGF-1 and epidermal growth factor-induced phospho-Ser-473-Akt, reduced Akt kinase activity, and blocked IGF-1 protection from UVC-induced apoptosis. The inhibition of Akt activity by TPA was reduced by inhibitors of protein phosphatase 2A, and TPA stimulated the association of phosphatase 2A with Akt. Individual PKC isoforms were overexpressed in cultured keratinocytes by transduction with adenoviral vectors or inhibited with PKC-selective inhibitors. These studies indicated that PKCδ and PKCϵ were selectively potent at causing dephosphorylation of Akt and modifying cell survival, whereas PKCα enhanced phosphorylation of Akt on Ser-473. Our results suggested that activation of PKCδ and PKCϵ provide a negative regulation for Akt phosphorylation and kinase activity in mouse keratinocytes and serve as modulators of cell survival pathways in response to external stimuli.


Journal of Cell Science | 2007

Reduced migration, altered matrix and enhanced TGFbeta1 signaling are signatures of mouse keratinocytes lacking Sdc1.

Mary Ann Stepp; Yueyuan Liu; Sonali Pal-Ghosh; Rosalyn A. Jurjus; Gauri Tadvalkar; Adith Sekaran; Kristen LoSicco; Li Jiang; Melinda Larsen; Luowei Li; Stuart H. Yuspa

We have reported previously that syndecan-1 (Sdc1)-null mice show delayed re-epithelialization after skin and corneal wounding. Here, we show that primary keratinocytes obtained from Sdc1-null mice and grown for 3-5 days in culture are more proliferative, more adherent and migrate more slowly than wt keratinocytes. However, the migration rates of Sdc1-null keratinocytes can be restored to wild-type levels by replating Sdc1-null keratinocytes onto tissue culture plates coated with fibronectin and collagen I, laminin (LN)-332 or onto the matrices produced by wild-type cells. Migration rates can also be restored by treating Sdc1-null keratinocytes with antibodies that block α6 or αv integrin function, or with TGFβ1. Antagonizing either β1 integrin function using a function-blocking antibody or TGFβ1 using a neutralizing antibody reduced wild-type keratinocyte migration more than Sdc1-null keratinocyte migration. Cultures of Sdc1-null keratinocytes accumulated less collagen than wild-type cultures but their matrices contained the same amount of LN-332. The Sdc1-null keratinocytes expressed similar total amounts of eight different integrin subunits but showed increased surface expression of αvβ6, αvβ8, and α6β4 integrins compared with wild-type keratinocytes. Whereas wild-type keratinocytes increased their surface expression of α2β1, αvβ6, αvβ8, and α6β4 after treatment with TGFβ1, Sdc1-null keratinocytes did not. Additional data from a dual-reporter assay and quantification of phosphorylated Smad2 show that TGFβ1 signaling is constitutively elevated in Sdc1-null keratinocytes. Thus, our results identify TGFβ1 signaling and Sdc1 expression as important factors regulating integrin surface expression, activity and migration in keratinocyte and provide new insight into the functions regulated by Sdc1.


Journal of Cell Science | 2007

CLIC4 mediates and is required for Ca2+-induced keratinocyte differentiation

Kwang S. Suh; Michihiro Mutoh; Tomoko Mutoh; Luowei Li; Andrew Ryscavage; John M. Crutchley; Rebecca A. Dumont; Christina Cheng; Stuart H. Yuspa

Keratinocyte differentiation requires integrating signaling among intracellular ionic changes, kinase cascades, sequential gene expression, cell cycle arrest, and programmed cell death. We now show that Cl– intracellular channel 4 (CLIC4) expression is increased in both mouse and human keratinocytes undergoing differentiation induced by Ca2+, serum and the protein kinase C (PKC)-activator, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Elevation of CLIC4 is associated with signaling by PKCδ, and knockdown of CLIC4 protein by antisense or shRNA prevents Ca2+-induced keratin 1, keratin 10 and filaggrin expression and cell cycle arrest in differentiating keratinocytes. CLIC4 is cytoplasmic in actively proliferating keratinocytes in vitro, but the cytoplasmic CLIC4 translocates to the nucleus in keratinocytes undergoing growth arrest by differentiation, senescence or transforming growth factor β (TGFβ) treatment. Targeting CLIC4 to the nucleus of keratinocytes via adenoviral transduction increases nuclear Cl– content and enhances expression of differentiation markers in the absence of elevated Ca2+. In vivo, CLIC4 is localized to the epidermis in mouse and human skin, where it is predominantly nuclear in quiescent cells. These results suggest that CLIC4 participates in epidermal homeostasis through both alterations in the level of expression and subcellular localization. Nuclear CLIC4, possibly by altering the Cl– and pH of the nucleus, contributes to cell cycle arrest and the specific gene expression program associated with keratinocyte terminal differentiation.


Cancer Research | 2010

RasGRP3 Contributes to Formation and Maintenance of the Prostate Cancer Phenotype

Dazhi Yang; Noemi Kedei; Luowei Li; Juan Tao; Julia F. Velasquez; Aleksandra M. Michalowski; Balázs István Tóth; Rita Marincsák; Attila Varga; Tamás Bíró; Stuart H. Yuspa; Peter M. Blumberg

RasGRP3 mediates the activation of the Ras signaling pathway that is present in many human cancers. Here, we explored the involvement of RasGRP3 in the formation and maintenance of the prostate cancer phenotype. RasGRP3 expression was elevated in multiple human prostate tumor tissue samples and in the human androgen-independent prostate cancer cell lines PC-3 and DU 145 compared with the androgen-dependent prostate cancer cell line LNCaP. Downregulation of endogenous RasGRP3 in PC-3 and DU 145 cells reduced Ras-GTP formation, inhibited cell proliferation, impeded cell migration, and induced apoptosis. Anchorage-independent growth of the PC-3 cells and tumor formation in mouse xenografts of both cell lines were likewise inhibited. Inhibition of RasGRP3 expression reduced AKT and extracellular signal-regulated kinase 1/2 phosphorylation and sensitized the cells to killing by carboplatin. Conversely, exogenous RasGRP3 elevated Ras-GTP, stimulated proliferation, and provided resistance to phorbol 12-myristate 13-acetate-induced apoptosis in LNCaP cells. RasGRP3-overexpressing LNCaP cells displayed a markedly enhanced rate of xenograft tumor formation in both male and female mice compared with the parental line. Suppression of RasGRP3 expression in these cells inhibited downstream RasGRP3 responses, caused the cells to resume the LNCaP morphology, and suppressed growth, confirming the functional role of RasGRP3 in the altered behavior of these cells. We conclude that RasGRP3 contributes to the malignant phenotype of the prostate cancer cells and may constitute a novel therapeutic target for human prostate cancer.


Oncogene | 2011

RasGRP3, a Ras activator, contributes to signaling and the tumorigenic phenotype in human melanoma

Dazhi Yang; Juan Tao; Luowei Li; Noemi Kedei; Zsuzsanna Tóth; Alexandra Czap; Julia F. Velasquez; Daniela Mihova; Aleksandra M. Michalowski; Stuart H. Yuspa; Peter M. Blumberg

RasGRP3, an activator for H-Ras, R-Ras and Ras-associated protein-1/2, has emerged as an important mediator of signaling downstream from receptor coupled phosphoinositide turnover in B and T cells. Here, we report that RasGRP3 showed a high level of expression in multiple human melanoma cell lines as well as in a subset of human melanoma tissue samples. Suppression of endogenous RasGRP3 expression in these melanoma cell lines reduced Ras-GTP formation as well as c-Met expression and Akt phosphorylation downstream from hepatocyte growth factor (HGF) or epidermal growth factor (EGF) stimulation. RasGRP3 suppression also inhibited cell proliferation and reduced both colony formation in soft agar and xenograft tumor growth in immunodeficient mice, demonstrating the importance of RasGRP3 for the transformed phenotype of the melanoma cells. Reciprocally, overexpression of RasGRP3 in human primary melanocytes altered cellular morphology, markedly enhanced cell proliferation and rendered the cells tumorigenic in a mouse xenograft model. Suppression of RasGRP3 expression in these cells inhibited downstream RasGRP3 responses and suppressed cell growth, confirming the functional role of RasGRP3 in the altered behavior of these cells. The identification of the role of RasGRP3 in melanoma highlights its importance, as a Ras activator, in the phosphoinositide signaling pathway in human melanoma and provides a new potential therapeutic target.


PLOS ONE | 2013

Screening Compounds with a Novel High-Throughput ABCB1-Mediated Efflux Assay Identifies Drugs with Known Therapeutic Targets at Risk for Multidrug Resistance Interference

Megan R. Ansbro; Suneet Shukla; Suresh V. Ambudkar; Stuart H. Yuspa; Luowei Li

ABCB1, also known as P-glycoprotein (P-gp) or multidrug resistance protein 1 (MDR1), is a membrane-associated multidrug transporter of the ATP-binding cassette (ABC) transporter family. It is one of the most widely studied transporters that enable cancer cells to develop drug resistance. Reliable high-throughput assays that can identify compounds that interact with ABCB1 are crucial for developing new therapeutic drugs. A high-throughput assay for measuring ABCB1-mediated calcein AM efflux was developed using a fluorescent and phase-contrast live cell imaging system. This assay demonstrated the time- and dose-dependent accumulation of fluorescent calcein in ABCB1-overexpressing KB-V1 cells. Validation of the assay was performed with known ABCB1 inhibitors, XR9576, verapamil, and cyclosporin A, all of which displayed dose-dependent inhibition of ABCB1-mediated calcein AM efflux in this assay. Phase-contrast and fluorescent images taken by the imaging system provided additional opportunities for evaluating compounds that are cytotoxic or produce false positive signals. Compounds with known therapeutic targets and a kinase inhibitor library were screened. The assay identified multiple agents as inhibitors of ABCB1-mediated efflux and is highly reproducible. Among compounds identified as ABCB1 inhibitors, BEZ235, BI 2536, IKK 16, and ispinesib were further evaluated. The four compounds inhibited calcein AM efflux in a dose-dependent manner and were also active in the flow cytometry-based calcein AM efflux assay. BEZ235, BI 2536, and IKK 16 also successfully inhibited the labeling of ABCB1 with radiolabeled photoaffinity substrate [125I]iodoarylazidoprazosin. Inhibition of ABCB1 with XR9576 and cyclosporin A enhanced the cytotoxicity of BI 2536 to ABCB1-overexpressing cancer cells, HCT-15-Pgp, and decreased the IC50 value of BI 2536 by several orders of magnitude. This efficient, reliable, and simple high-throughput assay has identified ABCB1 substrates/inhibitors that may influence drug potency or drug-drug interactions and predict multidrug resistance in clinical treatment.


Cancer Research | 2013

Abstract 5635: Development of a high-throughput cell and fluorescent image-based ABCB1-mediated efflux assay for screening inhibitors of ABCB1.

Megan R. Ansbro; Suneet Shukla; Suresh V. Ambudkar; Stuart H. Yuspa; Luowei Li

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC ABCB1, also known as P-glycoprotein (P-gp) or multidrug resistance protein 1 (MDR1), is a membrane-associated multidrug transporter of the ATP-binding cassette (ABC) transporter family. It is one of the most widely studied transporters that enable cancer cells to develop drug resistance. Reliable high-throughput assays that can identify compounds that interact with ABCB1 are crucial for developing new therapeutic drugs. A high-throughput assay for measuring ABCB1-mediated calcein AM efflux was developed using the IncuCyteFLR technology. Time- and dose-dependent accumulation of fluorescent calcein in ABCB1-overexpressing KB-V1 cells was recorded using the IncuCyteFLR. Validation of the assay was performed with known ABCB1 inhibitors, XR9576, verapamil, and cyclosporin A, all of which displayed dose-dependent inhibition of ABCB1-mediated calcein AM efflux in this assay. A kinase inhibitor library and other compounds with known therapeutic targets were screened. The assay is highly reproducible and inhibitors for ABCB1-mediated efflux were identified from the kinase inhibitor library. Among compounds with known targets, BEZ235, BI 2536, IKK 16, and Ispinesib (SB-715992) inhibited calcein AM efflux in a dose-dependent manner and were also active in the flow cytometry based efflux assay. BEZ235, BI 2536, and IKK 16 also successfully competed with radiolabeled photoaffinity substrate [125I]iodoarylazidoprazosin (IAAP) for binding to ABCB1. Inhibition of ABCB1 lowered the IC50 value of BI 2536 treated ABCB1-overexpressing cancer cells. Phase and fluorescent images taken by the IncuCyteFLR provided additional opportunities for evaluating compounds that are cytotoxic or produce false positive signals. This high-throughput assay provides an efficient, reliable, and simple technique for screening libraries of natural and synthetic compounds to identify ABCB1 inhibitors. The same approach may be applied to screen inhibitors of other ABC transporters when suitable cell lines and fluorescent substrates are used. Citation Format: Megan R. Ansbro, Suneet Shukla, Suresh V. Ambudkar, Stuart H. Yuspa, Luowei Li. Development of a high-throughput cell and fluorescent image-based ABCB1-mediated efflux assay for screening inhibitors of ABCB1. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5635. doi:10.1158/1538-7445.AM2013-5635


Cancer Research | 2010

Abstract 5029: RasGRP3- a new therapeutic target in human melanoma

Dazhi Yang; Tao Juan; Noemi Kedei; Luowei Li; Stuart H. Yuspa; Peter M. Blumberg

RasGRP3, a RasGEF (Ras guanine nucleotide exchange factor), is an activator of H-Ras, R-ras and Rap1, Here, we report that its expression level was increased in multiple human melanoma cell lines, reaching a level of protein expression approaching that of Ramos cells in the case of the SK-MEL-5 melanoma line. Likewise, RasGRP3 expression was variably elevated in human melanoma tissue samples. The importance of RasGRP3 expression for the melanoma cell lines was demonstrated by down regulating its expression, which inhibited cell proliferation and blocked both colony formation in soft agar and xenograft tumor formation in immunodeficient mice in the case of both the M14 and SK-Mel-5 cell lines. Finally, RasGRP3 was involved in downstream signaling in these cells. Suppression of RasGRP3 expression reduced both basal and HGF induced AKT phosphorylation in the M14 and SK-Mel-5 cells. Consistent with these results, we found that overexpression of RasGRP3 in human primary melanocytes changed their morphology, enhanced cell proliferation, and caused xenograft tumor formation. Suppression of the RasGRP3 overexpression in these cells inhibited downstream RasGRP3 responses and suppressed cell growth, confirming the functional role of RasGRP3 in the altered behavior of these cells. We conclude that RasGRP3 represents a potential therapeutic target in melanoma. Even more generally, the high level of expression and functional role of RasGRP3 in melanoma emphasizes that this RasGEF may play a more general role in regulation of Ras family members than had been initially appreciated. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5029.

Collaboration


Dive into the Luowei Li's collaboration.

Top Co-Authors

Avatar

Stuart H. Yuspa

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter M. Blumberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Henry Hennings

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suneet Shukla

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Suresh V. Ambudkar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noemi Kedei

University of Debrecen

View shared research outputs
Researchain Logo
Decentralizing Knowledge