Network


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

Hotspot


Dive into the research topics where Paul J. Davis is active.

Publication


Featured researches published by Paul J. Davis.


Annual Review of Pharmacology and Toxicology | 2011

Membrane Receptor for Thyroid Hormone: Physiologic and Pharmacologic Implications

Paul J. Davis; Faith B. Davis; Shaker A. Mousa; Mary K. Luidens; Hung Yun Lin

Plasma membrane integrin αvβ3 is a cell surface receptor for thyroid hormone at which nongenomic actions are initiated. L-thyroxine (T₄) and 3,3,5-triiodo-L-thyronine (T₃) promote angiogenesis and tumor cell proliferation via the receptor. Tetraiodothyroacetic acid (tetrac), a deaminated T₄ derivative, blocks the nongenomic proliferative and proangiogenic actions of T₄ and T₃. Acting at the integrin independently of T₄ and T₃, tetrac and a novel nanoparticulate formulation of tetrac that acts exclusively at the cell surface have oncologically desirable antiproliferative actions on multiple tumor cell survival pathway genes. These agents also block the angiogenic activity of vascular growth factors. Volume and vascular support of xenografts of human pancreatic, kidney, lung, and breast cancers are downregulated by tetrac formulations. The integrin αvβ3 receptor site for thyroid hormone selectively regulates signal transduction pathways and distinguishes between unmodified tetrac and the nanoparticulate formulation. The receptor also mediates nongenomic thyroid hormone effects on plasma membrane ion transporters and on intracellular protein trafficking.


Cell Cycle | 2009

Modification of survival pathway gene expression in human breast cancer cells by tetraiodothyroacetic acid (tetrac)

Anna B. Glinskii; Gennadi V. Glinsky; Hung Yun Lin; Heng Yuan Tang; Mingzeng Sun; Faith B. Davis; Mary K. Luidens; Shaker A. Mousa; Aleck H. Hercbergs; Paul J. Davis

Tetraiodothyroacetic acid (tetrac) inhibits the cellular actions of thyroid hormone initiated at the hormone receptor on plasma membrane integrin αvβ3. Via interaction with the integrin, tetrac is also capable of inhibiting the angiogenic effects of vascular endothelial growth factor and basic fibroblast growth factor. MDA-MB-231 cells are estrogen receptor-negative human breast cancer cells shown to be responsive to tetrac in terms of decreased cell proliferation. Here we describe actions initiated at the cell surface receptor by unmodified tetrac and nanoparticulate tetrac on a panel of survival pathway genes in estrogen receptor-negative human breast cancer (MDA-MB-231) cells. Nanoparticulate tetrac is excluded from the cell interior. Expression of apoptosis inhibitors XIAP (X-linked inhibitor of apoptosis) and MCL1 (myeloid cell leukemia sequence 1) was downregulated by nanoparticulate tetrac in these breast cancer cells whereas apoptosis-promoting CASP2 and BCL2L14 were upregulated by the nanoparticulate formulation. Unmodified tetrac affected only XIAP expression. Expression of the angiogenesis inhibitor thrombospondin 1 (THBS1) gene was increased by both formulations of tetrac, as was the expression of CBY1, a nuclear inhibitor of catenin activity. The majority of differentially regulated Ras-oncogene family members were downregulated by nanoparticulate tetrac. The latter downregulated expression of epidermal growth factor receptor gene and unmodified tetrac did not. Nanoparticulate tetrac has coherent anti-cancer actions on expression of differentially-regulated genes important to survival of MDA-MB-231 cells.


Steroids | 2012

Nongenomic effects of thyroid hormones on the immune system cells: New targets, old players.

Paolo De Vito; Valentina Balducci; Stefano Leone; Zulema A. Percario; Giorgio Mangino; Paul J. Davis; Faith B. Davis; Elisabetta Affabris; Paolo Luly; Jens Z. Pedersen; Sandra Incerpi

It is now widely accepted that thyroid hormones, l-thyroxine (T(4)) and 3,3,5-triiodo-l-thyronine (T(3)), act as modulators of the immune response. Immune functions such as chemotaxis, phagocytosis, generation of reactive oxygen species, and cytokine synthesis and release, are altered in hypo- and hyper-thyroid conditions, even though for many immune cells no clear correlation has been found between altered levels of T(3) or T(4) and effects on the immune responses. Integrins are extracellular matrix proteins that are important modulators of many cellular responses, and the integrin αvβ3 has been identified as a cell surface receptor for thyroid hormones. Rapid signaling via this plasma membrane binding site appears to be responsible for many nongenomic effects of thyroid hormones, independent of the classic nuclear receptors. Through the integrin αvβ3 receptor the hormone can activate both the ERK1/2 and phosphatidylinositol 3-kinase pathways, with downstream effects including intracellular protein trafficking, angiogenesis and tumor cell proliferation. It has recently become clear that an important downstream target of the thyroid hormone nongenomic pathway may be the mammalian target of rapamycin, mTOR. New results demonstrate the capability of T(3) or T(4) to induce in the short time range important responses related to the immune function, such as reactive oxygen species production and cell migration in THP-1 monocytes. Thus thyroid hormones seem to be able to modulate the immune system by a combination of rapid nongenomic responses interacting with the classical nuclear response.


Physiology & Behavior | 2010

Mini-review: Cell surface receptor for thyroid hormone and nongenomic regulation of ion fluxes in excitable cells

Paul J. Davis; Min Zhou; Faith B. Davis; Larry Lansing; Shaker A. Mousa; Hung Yun Lin

Thyroid hormone has been shown experimentally to affect cellular ion fluxes. For example, thyroid hormone-induced modulation has been described of cellular sodium current (I(Na)), inward rectifying potassium current (IKir) and sodium pump (Na, K-ATPase) and of calcium pump (Ca(2+)-ATPase) activities. Certain of these actions appear to reflect nongenomic mechanisms of hormone action that are initiated at the plasma membrane receptor for iodothyronines described on integrin alphavbeta3. One such action is the recent demonstration of enhancement by the hormone of I(Na) in neurons. Nongenomic actions of thyroid hormone initiated at the plasma membrane may be specifically inhibited by tetraiodothyroacetic acid (tetrac), a deaminated thyroid hormone analogue. Important behavioral changes are associated with clinical states of excessive or deficient thyroid function. The molecular basis for these changes has not been established. It is proposed that nongenomic actions of thyroid hormone in neurons-such as that on sodium current-underlie certain of these behaviors. The contribution of such nongenomic actions of the hormone to animal behavioral paradigms possibly relevant to thyroid hormone actions in human subjects may be tested in vivo with tetrac.


Steroids | 2016

Actions of l-thyroxine and Nano-diamino-tetrac (Nanotetrac) on PD-L1 in cancer cells

Hung Yun Lin; Yu-Tang Chin; André Wendindondé Nana; Ya-Jung Shih; Hsuan-Yu Lai; Heng-Yuan Tang; Matthew C. Leinung; Shaker A. Mousa; Paul J. Davis

The PD-1 (programmed death-1)/PD-L1 (PD-ligand 1) checkpoint is a critical regulator of activated T cell-cancer cell interactions, defending tumor cells against immune destruction. Nano-diamino-tetrac (NDAT; Nanotetrac) is an anticancer/anti-angiogenic agent targeted to the thyroid hormone-tetrac receptor on the extracellular domain of integrin αvβ3. NDAT inhibits the cancer cell PI3-K and MAPK signal transduction pathways that are critical to PD-L1 gene expression. We examined actions in vitro of thyroid hormone (l-thyroxine, T4) and NDAT on PD-L1 mRNA abundance (qPCR) and PD-L1 protein content in human breast cancer (MDA-MB-231) cells and colon carcinoma (HCT116 and HT-29) cells. In MDA-MB-231 cells, a physiological concentration of T4 (10-7M total; 10-10M free hormone) stimulated PD-L1 gene expression by 38% and increased PD-L1 protein by 2.7-fold (p<0.05, all changes). NDAT (10-7M) reduced PD-L1 in T4-exposed cells by 21% (mRNA) and 39% (protein) (p<0.05, all changes). In HCT116 cells, T4 enhanced PD-L1 gene expression by 17% and protein content by 24% (p<0.05). NDAT reduced basal PD-L1 mRNA by 35% and protein by 31% and in T4-treated cells lowered mRNA by 33% and protein by 66%. In HT-29 cells, T4 increased PD-L1 mRNA by 62% and protein by 27%. NDAT lowered basal and T4-stimulated responses in PD-L1 mRNA and protein by 35-40% (p<0.05). Activation of ERK1/2 was involved in T4-induced PD-L1 accumulation. We propose that, by a nongenomic mechanism, endogenous T4 may clinically support activity of the defensive PD-1/PD-L1 checkpoint in tumor cells. NDAT non-immunologically suppresses basal and T4-induced PD-L1 gene expression and protein accumulation in cancer cells.


Endocrine Research | 2013

Molecular Mechanisms of Actions of Formulations of the Thyroid Hormone Analogue, Tetrac, on the Inflammatory Response

Paul J. Davis; Gennadi V. Glinsky; Hung Yun Lin; Sandra Incerpi; Faith B. Davis; Shaker A. Mousa; Heng Yuan Tang; Aleck Hercbergs; Mary K. Luidens

Background. Tetraiodothyroacetic acid (tetrac) and its nanoparticulate formulation (Nanotetrac) act at a cell surface receptor to block angiogenesis and tumor cell proliferation. Objective. The complex anti-angiogenic properties of tetrac and Nanotetrac caused us to search in the literature and in certain of our unpublished mRNA experiments for evidence that these agents affect the early inflammatory response, perhaps through actions on specific cytokines and chemokines. Results and Discussion. Tetrac and Nanotetrac inhibit expression in tumor cells of cytokine genes, e.g., specific interleukins, and chemokine genes, such as fractalkine (CX3CL1), and chemokine receptor genes (CX3CR1) that have been identified as high priority targets in the development of inflammation-suppressant drugs. The possibility is also examined that tetrac formulations have an effect on the function of inflammatory cells.


Journal of Biomedical Science | 2017

Leptin OB3 peptide suppresses leptin-induced signaling and progression in ovarian cancer cells

Yu Tang Chin; Le Ming Wang; Meng Ti Hsieh; Ya Jung Shih; André Wendindondé Nana; Chun A. Changou; Yu Chen S.H. Yang; Hsien Chung Chiu; Earl Fu; Paul J. Davis; Heng Yuan Tang; Hung Yun Lin

BackgroundObesity and its comorbidities constitute a serious health burden worldwide. Leptin plays an important role in diet control; however, it has a stimulatory potential on cancer cell proliferation. The OB3 peptide, a synthetic peptide, was shown to be more active than leptin in regulating metabolism but with no mitogenic effects in cancer cells.MethodsIn this study, we investigated the proliferative effects, gene expressions and signaling pathways modulated by leptin and OB3 in human ovarian cancer cells. In addition, an animal study was performed.ResultsLeptin, but not OB3, induced the proliferation of ovarian cancer cells. Interestingly, OB3 blocked the leptin-induced proliferative effect when it was co-applied with leptin. Both leptin and OB3 activated the phosphatidylinositol-3-kinase (PI3K) signal transduction pathway. In addition, leptin stimulated the phosphorylation of signal transducer and activator of transcription-3 (STAT3) Tyr-705 as well as estrogen receptor (ER)α, and the expression of ERα-responsive genes. Interestingly, all leptin-induced signal activation and gene expressions were blocked by the co-incubation with OB3 and the inhibition of extracellular signal-regulated kinase (ERK)1/2. Coincidently, leptin, but not OB3, increased circulating levels of follicle-stimulating hormone (FSH) which is known to play important roles in the initiation and proliferation of ovarian cancer cells.ConclusionsIn summary, our findings suggest that the OB3 peptide may prevent leptin-induced ovarian cancer initiation and progression by disrupting leptin-induced proliferative signals via STAT3 phosphorylation and ERα activation. Therefore, the OB3 peptide is a potential anticancer agent that might be employed to prevent leptin-induced cancers in obese people.


Cell Cycle | 2017

In vitro effects of tetraiodothyroacetic acid combined with X-irradiation on basal cell carcinoma cells

John T. Leith; Paul J. Davis; Shaker A. Mousa; Aleck Hercbergs

ABSTRACT We investigated radiosensitization in an untreated basal cell carcinoma (TE.354.T) cell line and post-pretreatment with tetraiodothyroacetic acid (tetrac) X 1 h at 37°C, 0.2 and 2.0 µM tetrac. Radioresistant TE.354.T cells were grown in modified medium containing fibroblast growth factor-2, stem cell factor-1 and a reduced calcium level. We also added reproductively inactivated (30 Gy) “feeder cells” to the medium. The in vitro doubling time was 34.1 h, and the colony forming efficiency was 5.09 percent. These results were therefore suitable for clonogenic radiation survival assessment. The 250 kVp X-ray survival curve of control TE.354.T cells showed linear-quadratic survival parameters of αX-ray = 0.201 Gy−1 and βX-ray = 0.125 Gy−2. Tetrac concentrations of either 0.2 or 2.0 µM produced αX-ray and βX-ray parameters of 2.010 and 0.282 Gy−1 and 2.050 and 0.837 Gy−2, respectively. The surviving fraction at 2 Gy (SF2) for control cells was 0.581, while values for 0.2 and 2.0 µM tetrac were 0.281 and 0.024. The SF2 data show that tetrac concentrations of 0.2 and 2.0 µM sensitize otherwise radioresistant TE.354.T cells by factors of 2.1 and 24.0, respectively. Thus, radioresistant basal cell carcinoma cells may be radiosensitized pharmacologically by exposure to tetrac.


Endocrine Research | 2014

They live in the land down under: thyroid function and basal metabolic rate in the Blind Mole Rat, Spalax.

Aaron Avivi; Eviatar Nevo; Keren Cohen; Nick Sotnichenko; Aleck Hercbergs; Mark Band; Paul J. Davis; Martin Ellis; Osnat Ashur-Fabian

Abstract The Israeli blind subterranean mole rat (Spalax ehrenbergi superspecies) lives in sealed underground burrows under extreme, hypoxic conditions. The four Israeli Spalax allospecies have adapted to different climates, the cool–humid (Spalax galili, 2u2009nu2009=u200952 chromosomes), semihumid (S. golani, 2u2009nu2009=u200954) north regions, warm–humid (S. carmeli, 2u2009nu2009=u200958) central region and the warm–dry S. judaei, 2u2009nu2009=u200960) southern regions. A dramatic interspecies decline in basal metabolic rate (BMR) from north to south, even after years of captivity, indicates a genetic basis for this BMR trait. We examined the possibility that the genetically-conditioned interspecies BMR difference was expressed via circulating thyroid hormone. An unexpected north to south increase in serum free thyroxine (FT4) and total 3, 5, 3′-triiodo-L-thyronine (T3) (pu2009<u20090.02) correlated negatively with previously published BMR measurements. The increases in serum FT4 and T3 were symmetrical, so that the T3:FT4 ratio – interpretable as an index of conversion of T4 to T3 in nonthyroidal tissues – did not support relative decrease in production of T3 as a contributor to BMR. Increased north-to-south serum FT4 and T3 levels also correlated negatively with hemoglobin/hematocrit. North-to-south adaptations in spalacids include decreased BMR and hematocrit/hemoglobin in the face of increasing thyroid hormone levels, arguing for independent control of hormone secretion and BMR/hematocrit/hemoglobin. But the significant inverse relationship between thyroid hormone levels and BMR/hematocrit/hemoglobin is also consistent with a degree of cellular resistance to thyroid hormone action that protects against hormone-induced increase in oxygen consumption in a hostile, hypoxic environment.


Hormones and Cancer | 2018

Thyroid Hormone in the Clinic and Breast Cancer

Aleck Hercbergs; Shaker A. Mousa; Matthew C. Leinung; Hung Yun Lin; Paul J. Davis

There is preclinical and recent epidemiological evidence that thyroid hormone supports breast cancer. These observations raise the issue of whether management of breast cancer in certain settings should include consideration of reducing the possible contribution of thyroid hormone to the advancement of the disease. In a preliminary experience, elimination of the clinical action of endogenous L-thyroxine (T4) in patients with advanced solid tumors, including breast cancer, has favorably affected the course of the cancer, particularly when coupled with administration of exogenous 3,5,3′-triiodo-L-thyronine (T3) (euthyroid hypothyroxinemia). We discuss in the current brief review the possible clinical settings in which to consider whether endogenous thyroid hormone—or exogenous thyroid hormone in the patient with hypothyroidism and coincident breast cancer—is significantly contributing to breast cancer outcome.

Collaboration


Dive into the Paul J. Davis's collaboration.

Top Co-Authors

Avatar

Shaker A. Mousa

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Faith B. Davis

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hung Yun Lin

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Heng Yuan Tang

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Mary K. Luidens

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hung Yun Lin

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Gennadi V. Glinsky

Albany College of Pharmacy and Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge