Network


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

Hotspot


Dive into the research topics where Patricia S. Lorenzo is active.

Publication


Featured researches published by Patricia S. Lorenzo.


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

Phosphorylation of Protein Kinase Cδ on Distinct Tyrosine Residues Regulates Specific Cellular Functions

Ilana Kronfeld; Gila Kazimirsky; Patricia S. Lorenzo; Susan Garfield; Peter M. Blumberg; Chaya Brodie

Protein kinase Cδ (PKCδ) inhibits proliferation and decreases expression of the differentiation marker glutamine synthetase (GS) in C6 glioma cells. Here, we report that distinct, specific tyrosine residues on PKCδ are involved in these two responses. Transfection of cells with PKCδ mutated at tyrosine 155 to phenylalanine caused enhanced proliferation in response to 12-phorbol 12-myristate 13-acetate, whereas GS expression resembled that for the PKCδ wild-type transfectant. Conversely, transfection with PKCδ mutated at tyrosine 187 to phenylalanine resulted in increased expression of GS, whereas the rate of proliferation resembled that of the PKCδ wild-type transfectant. The tyrosine phosphorylation of PKCδ and the decrease in GS expression induced by platelet-derived growth factor (PDGF) were abolished by the Src kinase inhibitors PP1 and PP2. In response to PDGF, Fyn associated with PKCδ via tyrosine 187. Finally, overexpression of dominant negative Fyn abrogated the decrease in GS expression and reduced the tyrosine phosphorylation of PKCδ induced by PDGF. We conclude that the tyrosine phosphorylation of PKCδ and its association with tyrosine kinases may be an important point of divergence in PKC signaling.


Journal of Biological Chemistry | 1998

Protein Kinase C δ (PKCδ) Inhibits the Expression of Glutamine Synthetase in Glial Cells via the PKCδ Regulatory Domain and Its Tyrosine Phosphorylation

Chaya Brodie; Krisztina Bogi; Peter Acs; Patricia S. Lorenzo; Lindsey Baskin; Peter M. Blumberg

Protein kinase C (PKC) plays an important role in the proliferation and differentiation of glial cells. In a recent study we found that overexpression of PKCδ reduced the expression of the astrocytic marker glutamine synthetase (GS). In this study we explored the mechanisms involved in the inhibitory effect of PKCδ on the expression of glutamine synthetase. Using PKC chimeras we first examined the role of the catalytic and regulatory domains of PKCδ on the expression of glutamine synthetase. We found that cells stably transfected with chimeras between the regulatory domain of PKCδ and the catalytic domains of PKCα or ε inhibited the expression of GS, similar to the inhibition exerted by overexpression of PKCδ itself. In contrast, no significant effects were observed in cells transfected with the reciprocal PKC chimeras between the regulatory domains of PKCα or ε and the catalytic domain of PKCδ. PKCδ has been shown to undergo tyrosine phosphorylation in response to various activators. Tyrosine phosphorylation of PKCδ in response to phorbol 12-myristate 13-acetate and platelet-derived growth factor occurred only in chimeras which contained the PKCδ regulatory domain. Cells transfected with a PKCδ mutant (PKCδ5), in which the five putative tyrosine phosphorylation sites were mutated to phenylalanine, showed markedly diminished tyrosine phosphorylation in response to phorbol 12-myristate 13-acetate and platelet-derived growth factor and normal levels of GS. Our results indicate that the regulatory domain of PKCδ mediates the inhibitory effect of this isoform on the expression of GS. Phosphorylation of PKCδ on tyrosine residues in the regulatory domain is implicated in this inhibitory effect.


Cancer Research | 2007

Transgenic Overexpression of RasGRP1 in Mouse Epidermis Results in Spontaneous Tumors of the Skin

Carolyn E. Oki-Idouchi; Patricia S. Lorenzo

RasGRP1 is a guanine nucleotide exchange factor for Ras and a receptor of the second messenger diacylglycerol and its ultrapotent analogues, the phorbol esters. We have recently shown expression of RasGRP1 in the epidermal keratinocytes where it can mediate Ras activation in response to the phorbol ester 12-O-tetradecanoylphorbol-13-acetate, a well-known mouse skin tumor promoter. To explore the participation of RasGRP1 in skin carcinogenesis, we targeted the overexpression of RasGRP1 to basal epidermal keratinocytes using the keratin 5 promoter. These transgenic mice were viable and indistinguishable from their littermates, with normal differentiation and skin architecture. However, a percentage of the adult transgenic population developed spontaneous skin tumors, mainly squamous cell papillomas. The transgene was detected in the tumors as well as in primary keratinocytes isolated from transgenic mice. The transgenic keratinocytes also displayed elevated levels of active, GTP-loaded Ras compared with the levels observed in keratinocytes derived from wild-type littermates. We noticed a correlation between tumor incidence and wounding, which suggests that RasGRP1 overexpression may confer sensitivity to promotional stimuli, like wound repair mechanisms. Interestingly, we also found elevated levels of granulocyte colony-stimulating factor in conditioned media derived from transgenic keratinocytes subjected to in vitro wounding. Taken together, these data are the first to provide evidence of a novel role for RasGRP1 in skin carcinogenesis and suggest that RasGRP1 may participate in tumorigenesis through modulation of Ras and autocrine pathways.


Cancer Research | 2007

RasGRP1 overexpression in the epidermis of transgenic mice contributes to tumor progression during multistage skin carcinogenesis.

Courtney T. Luke; Carolyn E. Oki-Idouchi; J. Mark Cline; Patricia S. Lorenzo

RasGRP1 is a guanine nucleotide exchange factor for Ras, activated in response to the second messenger diacylglycerol and its ultrapotent analogues, the phorbol esters. We have previously shown that RasGRP1 is expressed in mouse epidermal keratinocytes and that transgenic mice overexpressing RasGRP1 in the epidermis under the keratin 5 promoter (K5.RasGRP1) are prone to developing spontaneous papillomas and squamous cell carcinomas, suggesting a role for RasGRP1 in skin tumorigenesis. Here, we examined the response of the K5.RasGRP1 mice to multistage skin carcinogenesis, using 7,12-dimethylbenz(a)anthracene as carcinogen and 12-O-tetradecanoylphorbol-13-acetate (TPA) as tumor promoter. We found that whereas tumor multiplicity did not differ between transgenic and wild-type groups, the transgenic tumors were significantly larger than those observed in the wild-type mice (wild-type, 4.58 +/- 0.25 mm; transgenic, 9.83 +/- 1.05 mm). Histologic analysis further revealed that squamous cell carcinomas generated in the transgenic mice were less differentiated and more invasive than the wild-type tumors. Additionally, 30% of the transgenic mice developed tumors in the absence of initiation, suggesting that RasGRP1 overexpression could partially substitute for the initiation step induced by dimethylbenz(a)anthracene. In primary keratinocytes isolated from K5.RasGRP1 mice, TPA stimulation induced higher levels of Ras activation compared with the levels measured in the wild-type cells, indicating that constitutive overexpression of RasGRP1 in epidermal cells leads to elevated biochemical activation of endogenous Ras in response to TPA. The present data suggests that RasGRP1 participates in skin carcinogenesis via biochemical activation of endogenous wild-type Ras and predisposes to malignant progression in cooperation with Ras oncogenic signals.


Journal of Biological Chemistry | 2006

The Exchange Factor and Diacylglycerol Receptor RasGRP3 Interacts with Dynein Light Chain 1 through Its C-terminal Domain

Sara M. Okamura; Carolyn E. Oki-Idouchi; Patricia S. Lorenzo

RasGRP3 is an exchange factor for Ras-like small GTPases that is activated in response to the second messenger diacylglycerol. As with other diacylglycerol receptors, RasGRP3 is redistributed upon diacylglycerol or phorbol ester binding. Several factors are important in determining the pattern of translocation, including the potency of the diacylglycerol analog, the affinity of the receptor for phospholipids, and in some cases, protein-protein interactions. However, little is known about the mechanisms that play a role in RasGRP3 redistribution aside from the nature of the ligand. To discover potential protein binding partners for RasGRP3, we screened a human brain cDNA library using a yeast two-hybrid approach. We identified dynein light chain 1 as a novel RasGRP3-interacting protein. The interaction was confirmed both in vitro and in vivo and required the C-terminal domain encompassing the last 127 amino acids of RasGRP3. A truncated mutant form of RasGRP3 that lacked this C-terminal domain was unable to interact with dynein light chain 1 and displayed a dramatically altered subcellular localization, with a strong reticular distribution and perinuclear and nuclear localization. These findings suggest that dynein light chain 1 represents a novel anchoring protein for RasGRP3 that may regulate subcellular localization of the exchange factor and, as such, may participate in the signaling mediated by diacylglycerol through RasGRP3.


American Journal of Pathology | 2009

RasGRP1 Transgenic Mice Develop Cutaneous Squamous Cell Carcinomas in Response to Skin Wounding : Potential Role of Granulocyte Colony-Stimulating Factor

Federico R. Diez; Ann A. Garrido; Amrish Sharma; Courtney T. Luke; James C. Stone; Nancy A. Dower; J. Mark Cline; Patricia S. Lorenzo

Models of epidermal carcinogenesis have demonstrated that Ras is a critical molecule involved in tumor initiation and progression. Previously, we have shown that RasGRP1 increases the susceptibility of mice to skin tumorigenesis when overexpressed in the epidermis by a transgenic approach, related to its ability to activate Ras. Moreover, RasGRP1 transgenic mice develop spontaneous papillomas and cutaneous squamous cell carcinomas, some of which appear to originate in sites of injury, suggesting that RasGRP1 may be responding to signals generated during the wound-healing process. In this study, we examined the response of the RasGRP1 transgenic animals to full-thickness incision wounding of the skin, and demonstrated that they respond by developing tumors along the wounded site. The tumors did not present mutations in the H-ras gene, but Rasgrp1 transgene dosage correlated with tumor susceptibility and size. Analysis of serum cytokines showed increased levels of granulocyte colony-stimulating factor in transgenic animals after wounding. Furthermore, in vitro experiments with primary keratinocytes showed that granulocyte colony-stimulating factor stimulated Ras activation, although RasGRP1 was dispensable for this effect. Since granulocyte colony-stimulating factor has been recently associated with proliferation of skin cancer cells, our results may help in the elucidation of pathways that activate Ras in the epidermis during tumorigenesis in the absence of oncogenic ras mutations.


Journal of Biological Chemistry | 2010

RasGRP1 Is Essential for Ras Activation by the Tumor Promoter 12-O-Tetradecanoylphorbol-13-acetate in Epidermal Keratinocytes

Amrish Sharma; Courtney T. Luke; Nancy A. Dower; James C. Stone; Patricia S. Lorenzo

RasGRP1 is a guanine nucleotide exchange factor for Ras that binds with high affinity to diacylglycerol analogs like the phorbol esters. Recently, we demonstrated a role for RasGRP1 in skin carcinogenesis and suggested its participation in the action of tumor-promoting phorbol esters like 12-O-tetradecanoylphorbol-13-acetate (TPA) on Ras pathways in epidermal cells. Given the importance of Ras in carcinogenesis, we sought to discern whether RasGRP1 was a critical pathway in Ras activation, using a RasGRP1 knockout (KO) mouse model to examine the response of keratinocytes to TPA. In contrast to the effect seen in wild type keratinocytes, RasGTP levels were barely detected in RasGRP1 KO cells even after 60 min of exposure to phorbol esters. The lack of response was rescued by enforced expression of RasGRP1. Furthermore, small hairpin RNA-induced silencing of RasGRP1 abrogated the effect of TPA on Ras. Analysis of Ras isoforms showed that both H-Ras and N-Ras depended on RasGRP1 for activation by TPA, whereas activation of K-Ras could not be detected. Although RasGRP1 was dispensable for ERK activation in response to TPA, JNK activation was reduced in the KO keratinocytes. Notably, TPA-induced phosphorylation of JNK2, but not JNK1, was reduced by RasGRP1 depletion. These data identify RasGRP1 as a critical molecule in the activation of Ras by TPA in primary mouse keratinocytes and suggest JNK2 as one of the relevant downstream targets. Given the role of TPA as a skin tumor promoter, our findings provide additional support for a role for RasGRP1 in skin carcinogenesis.


Archive | 2000

Inhibitors of Protein Kinase C and Related Receptors for the Lipophilic Second-Messenger sn-1,2-Diacylglycerol

Peter M. Blumberg; Peter Acs; Dipak K. Bhattacharyya; Patricia S. Lorenzo

Toxic natural products, selected by evolution both for activity against crucial biological targets and for potency, have made major contributions in highlighting such targets and in defining their functions. The phorbol esters, initially isolated on the basis of their activity as tumor promoters and as acute irritants, were found to have dramatic biological effects in virtually every system in which they were examined (1). We now appreciate that the phorbol esters function as ultrapotent analogs of the lipophilic second messenger sn-1,2-diacylglycerol (DAG) and that phorbol ester receptors identify high-affinity targets in DAG signaling (2), of which protein kinase C (PKC) has been most thoroughly studied (3–7).


Carcinogenesis | 2014

Targeted deletion of RasGRP1 impairs skin tumorigenesis

Amrish Sharma; Lauren L. Fonseca; Cynthia Rajani; Jodi K. Yanagida; Yuka Endo; J. Mark Cline; James C. Stone; Junfang Ji; Joe W. Ramos; Patricia S. Lorenzo

Ras is frequently activated in cutaneous squamous cell carcinoma, a prevalent form of skin cancer. However, the pathways that contribute to Ras-induced transformation have not been entirely elucidated. We have previously demonstrated that in transgenic mice, overexpression of the Ras activator RasGRP1 promotes the formation of spontaneous skin tumors and enhances malignant progression in the multistage carcinogenesis skin model that relies on the oncogenic activation of H-Ras. Utilizing a RasGRP1 knockout mouse model (RasGRP1 KO), we now show that lack of RasGRP1 reduced the susceptibility to skin tumorigenesis. The dependency on RasGRP1 was associated with a diminished response to the phorbol ester tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Specifically, we found impairment of epidermal hyperplasia induced by TPA through keratinocyte proliferation. Using a keratinocyte cell line that carries a ras oncogenic mutation, we also demonstrated that RasGRP1 could further activate Ras in response to TPA. Thus, we propose that RasGRP1 upregulates signaling from Ras and contributes to epidermal tumorigenesis by increasing the total dosage of active Ras.

Collaboration


Dive into the Patricia S. Lorenzo's collaboration.

Top Co-Authors

Avatar

Peter M. Blumberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amrish Sharma

University of Hawaii at Manoa

View shared research outputs
Top Co-Authors

Avatar

Courtney T. Luke

University of Hawaii at Manoa

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Krisztina Bogi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Acs

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge