Network


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

Hotspot


Dive into the research topics where Robert A. Sikes is active.

Publication


Featured researches published by Robert A. Sikes.


The Prostate | 2000

LNCaP progression model of human prostate cancer: Androgen‐independence and osseous metastasis

George N. Thalmann; Robert A. Sikes; Tony T. Wu; Armelle Degeorges; Shi-Ming Chang; Mustafa Ozen; Sen Pathak; Leland W. K. Chung

Clinically, the lethal phenotypes of human prostate cancer are characterized by their progression to androgen‐independence and their propensity to form osseous metastases. We reported previously on the establishment of androgen‐independent (AI) human prostate cancer cell lines derived from androgen‐dependent (AD) LNCaP cells, with androgen independence defined as the capability of prostate cancer cells to grow in castrated hosts. One of the sublines, C4‐2, was found to be AI, highly tumorigenic, and metastatic, having a proclivity for metastasis to the bone.


International Journal of Cancer | 1998

Establishing human prostate cancer cell xenografts in bone: Induction of osteoblastic reaction by prostate‐specific antigen‐producing tumors in athymic and SCID/bg mice using LNCaP and lineage‐derived metastatic sublines

Tony T. Wu; Robert A. Sikes; Quanjun Cui; George N. Thalmann; Chinghai Kao; Cheryl F. Murphy; Hua Yang; Haiyen E. Zhau; Gary Balian; Leland W. K. Chung

LNCaP lineage‐derived human prostate cancer cell lines C4‐2 and C4‐2B4 acquire androgen independence and osseous metastatic potential in vivo. Using C4‐2 and C4‐2B4 the goals of the current investigation were 1) to establish an ideal bone xenograft model for prostate cancer cells in intact athymic or SCID/bg mice using an intraosseous route of tumor cell administration and 2) to compare prostate cancer metastasis by administering cells either through intravenous (i.v.) or intracardiac administration in athymic or SCID/bg mice. Subsequent to tumor cell administration, prostate cancer growth in the skeleton was assessed by radiographic bone density, serum prostate‐specific antigen (PSA) levels, presence of hematogenous prostate cancer cells and histopathologic evaluation of tumor specimens in the lymph node and skeleton. Our results show that whereas LNCaP cells injected intracardially failed to develop metastasis, C4‐2 cells injected similarly had the highest metastatic capability in SCID/bg mice. Retroperitoneal and mediastinal lymph node metastases were noted in 3/7 animals, whereas 2/7 animals developed osteoblastic spine metastases. Intracardiac injection of C4‐2 in athymic hosts produced spinal metastases in 1/5 animals at 8–12 weeks post‐injection; PC‐3 injected intracardially also metastasized to the bone but yielded osteolytic responses. Intravenous injection of either LNCaP or C4‐2 failed to establish tumor colonies. Intrailiac injection of C4‐2 but not LNCaP nor C4‐2B4 cells in athymic mice established rapidly growing tumors in 4/8 animals at 2–7 weeks after inoculation. Intrafemoral injection of C4‐2 (9/16) and C4‐2B4 (5/18) but not LNCaP (0/13) cells resulted in the development of osteoblastic bone lesions in athymic mice (mean: 6 weeks, range: 3–12 weeks). In SCID/bg mice, intrafemoral injection of LNCaP (6/8), C4‐2 (8/8) and C4‐2B4 (8/8) cells formed PSA‐producing, osteoblastic tumors in the bone marrow space within 3–5 weeks after tumor cell inoculation. A stepwise increase of serum PSA was detected in all animals. Reverse transcription‐polymerase chain reaction (RT‐PCR) to detect hematogenously disseminated prostate cancer cells could not be correlated to either serum PSA level or histological evidence of tumor cells in the marrow space. We have thus established a PSA‐producing and osteoblastic human prostate cancer xenograft model in mice. Int. J. Cancer 77:887–894, 1998.© 1998 Wiley‐Liss, Inc.


Photochemistry and Photobiology | 1994

PHOTODYNAMIC THERAPY WITH PHOTOFRIN II INDUCES PROGRAMMED CELL DEATH IN CARCINOMA CELL LINES

Xiao-Yan He; Robert A. Sikes; Sharon L. Thomsen; Leland W.K. Chung; Steven L. Jacques

Abstract The mode of cell death following photodynamic therapy was investigated from the perspective of programmed cell death or apoptosis. Human prostate carcinoma cells (PC3), human non‐small cell lung carcinoma (H322a) and rat mammary carcinoma (MTF7) were treated by photodynamic therapy. An examination of extracted cellular DNA by gel electrophoresis showed the characteristic DNA ladder indicative of internucleosomal cleavage of DNA during apoptosis. The magnitude of the response and the photodynamic therapy dosage required to induce DNA fragmentation were different in PC3 and MTF7. The MTF7 cells responded with rapid apoptosis at the dose of light and drug that yielded 50% cell death (LD50). In contrast, PC3 showed only marginal response at the LD50 but had a marked response at the LD85. Thus, apoptosis did not ensue as quickly in PC3 as in MTF7. The H322a cells were killed by photodynamic therapy but failed to exhibit any apoptotic response. The results also suggested that apoptosis in these cell lines has a minor requirement for de novo protein synthesis and no requirement for de novo RNA synthesis. This study indicates that although apoptosis can occur during photodynamic therapy‐induced cell death, this response is not universal for all cancer cell lines.


Cancer Research | 2004

Increased Insulin-Like Growth Factor I Receptor Expression and Signaling Are Components of Androgen-Independent Progression in a Lineage-Derived Prostate Cancer Progression Model

Sandra L. Krueckl; Robert A. Sikes; N. Magnus Edlund; Robert H. Bell; Antonio Hurtado-Coll; Ladan Fazli; Martin Gleave; Michael E. Cox

Apoptosis and inhibition of mitosis are primary mechanisms mediating androgen ablation therapy-induced regression of prostate cancer (PCa). However, PCa readily becomes androgen independent, leading to fatal disease. Up-regulated growth and survival signaling is implicated in development of resistance to androgen ablation therapy. We are testing the hypothesis that insulin-like growth factor (IGF) responsiveness is required for androgen-independent (AI) progression. Using the LNCaP human PCa progression model, we have determined that IGF-I–mediated protection from apoptotic stress and enhanced mitotic activity is androgen dependent in LNCaP cells but is androgen independent in lineage-derived C4-2 cells. Both cell lines exhibit androgen-responsive patterns of IGF-I receptor (IGF-IR) expression, activation, and signaling to insulin receptor substrate-2 and AKT. However, C4-2 cells express higher levels of IGF-IR mRNA and protein and exhibit enhanced IGF-I–mediated phosphorylation and downstream signaling under androgen-deprived conditions. In comparisons of naïve and AI metastatic human PCa specimens, we have confirmed that IGF-IR levels are elevated in advanced disease. Together with our LNCaP/C4-2 AI progression model data, these results indicate that increased IGF-IR expression is associated with AI antiapoptotic and promitotic IGF signaling in PCa disease progression.


Reproductive Biology and Endocrinology | 2004

Changes in extracellular matrix (ECM) and ECM-associated proteins in the metastatic progression of prostate cancer

Delisha A Stewart; Carlton R. Cooper; Robert A. Sikes

Prostate cancer (PCa) is no exception to the multi-step process of metastasis. As PCa progresses, changes occur within the microenvironments of both the malignant cells and their targeted site of metastasis, enabling the necessary responses that result in successful translocation. The majority of patients with progressing prostate cancers develop skeletal metastases. Despite advancing efforts in early detection and management, there remains no effective, long-term cure for metastatic PCa. Therefore, the elucidation of the mechanism of PCa metastasis and preferential establishment of lesions in bone is an intensive area of investigation that promises to generate new targets for therapeutic intervention. This review will survey what is currently know concerning PCa interaction with the extracellular matrix (ECM) and the roles of factors within the tumor and ECM microenvironments that contribute to metastasis. These will be discussed within the context of changes in expression and functional heterodimerization patterns of integrins, changes in ECM expression and reorganization by proteases facilitating invasion. In this context we also provide a brief summary of how growth factors (GFs), cytokines and regulatory signaling pathways favor PCa metastasis to bone.


Cancer and Metastasis Reviews | 2006

Voltage-sensitive ion channels and cancer.

Jamie L. Fiske; Victor P. Fomin; Milton L. Brown; Randall L. Duncan; Robert A. Sikes

Plasma membrane voltage-sensitive ion channels classically have been associated with a variety of inherited diseases or “channelopathies” that range in the severity of symptoms from mild to lethal. Ion channels are found throughout the body and are responsible for facilitated diffusion of ions down the electrochemical gradient across cells membranes in various tissues. Voltage-sensitive ion channels open in response to changes in the membrane potential and are primarily found in excitable cells and tissues. Potassium, calcium, and sodium channels play critical roles in the development of major diseases, such as hyperkalemia, epilepsy, congenital myotonia and several cardiac arrythmias. Recently, cancer studies have begun to define the role of voltage-sensitive ion channels in the progression of cancer to a more malignant phenotype. In cancer, the increased expression or increased kinetics of voltage-sensitive ion channels is associated with an increasing malignant potential as evinced by their role in cell proliferation, migration and survival; as such, these channels are becoming the targets of significant drug development efforts to block or reduce voltage-sensitive ion channel activity in order to prevent or combat malignant disease.


Journal of Biological Chemistry | 1998

Isolation and characterization of PAGE-1 and GAGE-7. New genes expressed in the LNCaP prostate cancer progression model that share homology with melanoma-associated antigens.

Michael E. Chen; Sue Hwa Lin; Leland W. K. Chung; Robert A. Sikes

The LNCaP progression model of human prostate cancer consists of lineage-related sublines that differ in their androgen sensitivity and metastatic potential. A differential display polymerase chain reaction was employed to evaluate mRNA expression differences between the LNCaP sublines in order to define the differences in gene expression between the androgen-sensitive, nontumorigenic LNCaP cell line and the androgen-insensitive, metastatic LNCaP sublines, C4-2 and C4-2B. An amplicon, BG16.21, was isolated that showed increased expression in the androgen-independent and metastatic LNCaP sublines, C4-2 and C4-2B. Hybridization screening of a λ gt11 expression library with BG16.21 revealed two transcripts, both homologous to BG16.21 at the 3′ end. A GenBankTM data base search using the GCG Wisconsin software package revealed the shorter ∼600-bp transcript (designated GAGE-7) to be a new member of the GAGE family. The second ∼700-bp transcript was a novel gene (designated PAGE-1, “prostate associated gene”) with only 45% homology to GAGE gene family members. RNA blot analysis demonstrated that GAGE-7mRNA was expressed at equal levels in all lineage related prostate cancer cell sublines, while PAGE-1 mRNA levels were elevated 5-fold in C4-2 and C4-2B as compared with LNCaP cells. NeitherGAGE-7 nor PAGE-1 demonstrated any regulation by androgens in the prostate cancer cell lines used in this study.PAGE-1 and GAGE-7 expression was found to be restricted to testes (high) and placenta (low) on human multiple tissue Northern blots. As GAGE/MAGE antigens were reported previously to be targets for tumor-specific cytotoxic lymphocytes in melanoma, these results suggest that PAGE-1 and GAGE-7 may be related to prostate cancer progression and may serve as potential targets for novel therapies.


International Journal of Cancer | 2004

Cellular interactions in the tropism of prostate cancer to bone

Robert A. Sikes; Brian Nicholson; Kenneth S. Koeneman; N. Magnus Edlund; Eric A. Bissonette; Michael Bradley; George N. Thalmann; Marco G. Cecchini; Kenneth J. Pienta; Leland W.K. Chung

At autopsy ≥80% of prostate cancers have established macrometastases in marrow containing bone. The mechanism(s) to explain this remarkable level of bone involvement remain to be elucidated. We examined the adhesive and invasive behavior of prostate cancer cells to osteoblastic and human bone marrow endothelial cells (HBME‐1) in an attempt to explain the tropism of prostate cells for bone. We found an inverse relationship between adhesion and prostate cell tumorigenicity and metastatic potential. Relative cell adhesion of P69 between cell lines was 1.74‐fold (95% confidence interval [CI] = 1.15–2.64) and 1.58‐fold (95% CI = 0.94–2.68) greater at 1 hr and 2 hr, respectively, than LNCaP that was essentially equivalent to C4‐2 cells when using an osteoblastic cell line, D1 as the substrate. Similar results were acquired when HBME‐1 were used as substratum. There was a marked increase in adhesion of the poorly tumorigenic cell line P69 as compared to the cancer cells to HBME‐1. P69 adhesion was 2.78‐fold (95% CI = 1.87–4.84) and 2.0‐fold (95% CI = 1.43–2.80) greater at 1 hr and 2 hr, respectively when compared to LNCaP or C4‐2 cells. D1 cells, a bone homing osteoblastic precursor, behaved contrary to the metastatic, bone‐colonizing C4‐2 cell line and bound best to other bone cells but not as well as a non‐homing fetal bone marrow‐derived cell line, D2. Invasion of prostate cancer cells through HBME‐1 lawns was examined at 8 hr and 16 hr. In contrast to the adhesion studies, the invasion of the more aggressive C4‐2 cells was 3.46‐fold (95% CI = 1.18–10.17) and 2.65‐fold (95% CI = 1.26–5.56) greater at 8 hr and 16 hr, respectively than LNCaP cells. Similarly, LNCaP cell invasion was 1.73‐fold (95% CI = 0.69–4.37) and 2.35‐fold (95% CI = 1.41–3.93) greater at 8 hr and 16 hr, respectively than P69 cells at the invasion of HBME‐1 monolayers. At 8 hr, C4‐2 cells had 6.0‐fold (95% CI = 2.63,13.65) higher invasive potential than P69 cells. Phage display biopanning of LNCaP cells versus C4‐2 cells in vitro using 4 separate techniques repeatedly identified the same peptide in support of minimal cell surface changes associated with the ability of C4‐2 cells to metastasize to bone. As integrins are vital to cell adhesion and migration, we examined the integrin subunit expression in the prostate cell lines. The expression of integrin subunits is much higher in the nontumorigenic cell line, P69, whereas the differences in integrin expression between LNCaP and C4‐2 are negligible. Only α2 and β5 integrin subunits increase from LNCaP to C4‐2. Given that C4‐2 cells spontaneously metastasize to bone in vivo and LNCaP cells do not, these studies imply that the ability of a metastatic prostate cancer cell to colonize the bone is not completely dependent upon the ability of the cancer cell to adhere to either osteoblastic cells or to the bone marrow endothelial cell lining. Therefore, the initial interaction between the bone endothelium or stroma and prostate cells is not accurately referred to as a tropic or homing response. The invasion assay results indicate that the invasive potential of the cell more accurately reflects the bone colonizing potential of a prostate cancer cell. It is likely that bidirectional paracrine interactions, subsequent to marrow adhesion, between prostate cancer cells and the bone microenvironment are what determine the successful colonization of the bone by prostate cancer cells. Further, functional changes in surface proteins that are involved in invasion are likely to occur without major changes in levels of cell surface protein expression. Functional integrin association, substratum usage and outside in signaling are more likely to predict metastatic behavior.


Journal of Histochemistry and Cytochemistry | 2000

Distribution of IGFBP-rP1 in Normal Human Tissues

Armelle Degeorges; Fuan Wang; Henry F. Frierson; Arun Seth; Robert A. Sikes

IGFBP-rP1/mac25 is a recently described member of the insulin-like growth factor binding protein (IGFBP) family. It has structural homology to the other members of the IGFBP family but has a lower affinity for insulin-like growth factors (IGFs). In previous studies using RNA blot hybridization, it was shown that the expression of IGFBP-rP1/mac25 was ubiquitous in normal human tissues. In this report we show by immunohistochemistry that the expression of IGFBP-rP1/mac25 is actually restricted to certain organs and specific cell types. We used an antibody raised against a decapeptide of the C-terminal part of the protein that recognizes a ≍37-kD protein under reduced conditions. The immunohistochemistry performed on normal human tissues showed a ubiquitous intense staining of peripheral nerves and a variable degree of positive staining in smooth muscle cells, including those from blood vessel walls, gut, bladder, and prostate. Cilia from the respiratory system, epididymis, and fallopian tube showed intense immunoreactivity. Most endothelial cells showed some positivity, whereas fat cells, plasma cells, and lymphocytes were negative. There was specific expression limited to certain cell types in the kidney, adrenal gland, and skeletal muscle, indicating a possible specialized function of IGFBP-rP1/mac25 in these organs. We further noted an opposite pattern of staining in the lining epithelium of breast (typically positive) and prostate glands (largely negative). The specific localization of IGFBP-rP1/mac25 as described implies a function of the protein. However, its regulation within the IGF axis or a possible direct action of IGFBP-rP1/mac25 remains to be demonstrated.


BMC Cancer | 2006

Met-Independent Hepatocyte Growth Factor-mediated regulation of cell adhesion in human prostate cancer cells

Amanda W. Tate; Shuji Isotani; Michael Bradley; Robert A. Sikes; Rodney Davis; Leland W.K. Chung; Magnus Edlund

BackgroundProstate cancer cells communicate reciprocally with the stromal cells surrounding them, inside the prostate, and after metastasis, within the bone. Each tissue secretes factors for interpretation by the other. One stromally-derived factor, Hepatocyte Growth Factor (HGF), was found twenty years ago to regulate invasion and growth of carcinoma cells. Working with the LNCaP prostate cancer progression model, we found that these cells could respond to HGF stimulation, even in the absence of Met, the only known HGF receptor. The new HGF binding partner we find on the cell surface may help to clarify conflicts in the past literature about Met expression and HGF response in cancer cells.MethodsWe searched for Met or any HGF binding partner on the cells of the PC3 and LNCaP prostate cancer cell models, using HGF immobilized on agarose beads. By using mass spectrometry analyses and sequencing we have identified nucleolin protein as a novel HGF binding partner. Antibodies against nucleolin (or HGF) were able to ameliorate the stimulatory effects of HGF on met-negative prostate cancer cells. Western blots, RT-PCR, and immunohistochemistry were used to assess nucleolin levels during prostate cancer progression in both LNCaP and PC3 models.ResultsWe have identified HGF as a major signaling component of prostate stromal-conditioned media (SCM) and have implicated the protein nucleolin in HGF signal reception by the LNCaP model prostate cancer cells. Antibodies that silence either HGF (in SCM) or nucleolin (on the cell surfaces) eliminate the adhesion-stimulatory effects of the SCM. Likewise, addition of purified HGF to control media mimics the action of SCM. C4-2, an LNCaP lineage-derived, androgen-independent human prostate cancer cell line, responds to HGF in a concentration-dependent manner by increasing its adhesion and reducing its migration on laminin substratum. These HGF effects are not due to shifts in the expression levels of laminin-binding integrins, nor can they be linked to expression of the known HGF receptor Met, as neither LNCaP nor clonally-derived C4-2 sub-line contain any detectable Met protein. Even in the absence of Met, small GTPases are activated, linking HGF stimulation to membrane protrusion and integrin activation. Membrane-localized nucelolin levels increase during cancer progression, as modeled by both the PC3 and LNCaP prostate cancer progression cell lines.ConclusionWe propose that cell surface localized nucleolin protein may function in these cells as a novel HGF receptor. Membrane localized nucleolin binds heparin-bound growth factors (including HGF) and appears upregulated during prostate cancer progression. Antibodies against nucleolin are able to ameliorate the stimulatory effects of HGF on met-negative prostate cancer cells. HGF-nucleolin interactions could be partially responsible for the complexity of HGF responses and met expression reported in the literature.

Collaboration


Dive into the Robert A. Sikes's collaboration.

Top Co-Authors

Avatar

Leland W.K. Chung

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fayth L. Miles

University of California

View shared research outputs
Top Co-Authors

Avatar

David J. DeGraff

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Haiyen E. Zhau

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qian Chen

University of Delaware

View shared research outputs
Researchain Logo
Decentralizing Knowledge