Network


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

Hotspot


Dive into the research topics where Murat Yalcin is active.

Publication


Featured researches published by Murat Yalcin.


Nanotechnology | 2009

Gold and silver nanoparticles conjugated with heparin derivative possess anti-angiogenesis properties

Melissa M. Kemp; Ashavani Kumar; Shaymaa S. Mousa; Evgeny Dyskin; Murat Yalcin; Pulickel M. Ajayan; Robert J. Linhardt; Shaker A. Mousa

Silver and gold nanoparticles display unique physical and biological properties that have been extensively studied for biological and medical applications. Typically, gold and silver nanoparticles are prepared by chemical reductants that utilize excess toxic reactants, which need to be removed for biological purposes. We utilized a clean method involving a single synthetic step to prepare metal nanoparticles for evaluating potential effects on angiogenesis modulation. These nanoparticles were prepared by reducing silver nitrate and gold chloride with diaminopyridinyl (DAP)-derivatized heparin (HP) polysaccharides. Both gold and silver nanoparticles reduced with DAPHP exhibited effective inhibition of basic fibroblast growth factor (FGF-2)-induced angiogenesis, with an enhanced anti-angiogenesis efficacy with the conjugation to DAPHP (P<0.01) as compared to glucose conjugation. These results suggest that DAPHP-reduced silver nanoparticles and gold nanoparticles have potential in pathological angiogenesis accelerated disorders such as cancer and inflammatory diseases.


The Journal of Clinical Endocrinology and Metabolism | 2010

Tetraiodothyroacetic Acid (Tetrac) and Nanoparticulate Tetrac Arrest Growth of Medullary Carcinoma of the Thyroid

Murat Yalcin; Evgeny Dyskin; Lawrence Lansing; Dhruba J. Bharali; Shaymaa S. Mousa; A. Bridoux; Aleck H. Hercbergs; Hung-Yun Lin; Faith B. Davis; Gennadi V. Glinsky; A. Glinskii; J. Ma; Paul J. Davis; Shaker A. Mousa

CONTEXT Tetraiodothyroacetic acid (tetrac) blocks angiogenic and tumor cell proliferation actions of thyroid hormone initiated at the cell surface hormone receptor on integrin alphavbeta3. Tetrac also inhibits angiogenesis initiated by vascular endothelial growth factor and basic fibroblast growth factor. OBJECTIVE We tested antiangiogenic and antiproliferative efficacy of tetrac and tetrac nanoparticles (tetrac NP) against human medullary thyroid carcinoma (h-MTC) implants in the chick chorioallantoic membrane (CAM) and h-MTC xenografts in the nude mouse. DESIGN h-MTC cells were implanted in the CAM model (n = 8 per group); effects of tetrac and tetrac NP at 1 microg/CAM were determined on tumor angiogenesis and tumor growth after 8 d. h-MTC cells were also implanted sc in nude mice (n = 6 animals per group), and actions on established tumor growth of unmodified tetrac and tetrac NP ip were determined. RESULTS In the CAM, tetrac and tetrac NP inhibited tumor growth and tumor-associated angiogenesis. In the nude mouse xenograft model, established 450-500 mm(3) h-MTC tumors were reduced in size over 21 d by both tetrac formulations to less than the initial cell mass (100 mm(3)). Tumor tissue hemoglobin content of xenografts decreased by 66% over the course of administration of each drug. RNA microarray and quantitative real-time PCR of tumor cell mRNAs revealed that both tetrac formulations significantly induced antiangiogenic thrombospondin 1 and apoptosis activator gene expression. CONCLUSIONS Acting via a cell surface receptor, tetrac and tetrac NP inhibit growth of h-MTC cells and associated angiogenesis in CAM and mouse xenograft models.


Lung Cancer | 2012

Tetraiodothyroacetic acid and its nanoformulation inhibit thyroid hormone stimulation of non-small cell lung cancer cells in vitro and its growth in xenografts.

Shaker A. Mousa; Murat Yalcin; Dhruba J. Bharali; Ran Meng; Heng Yuan Tang; Hung Yun Lin; Faith B. Davis; Paul J. Davis

Thyroid hormone stimulates cell proliferation of several types of cancers and stimulates cancer-relevant angiogenesis. In the present study, we investigated the proliferative effect of thyroid hormone and the anti-proliferative and anti-angiogenic action of its nano-derivative, tetrac-NP, on human non-small cell lung cancer (NSCLC) H1299 cells in vitro and in xenografts. The anti-proliferative activity of unmodified tetrac and tetrac-NP against human H1299 cells was determined in three models: (a) cultured H1299 cells in vitro, (b) tumor cell implants in the fertilized chick chorioallantoic membrane (CAM) system and (c) xenografts in the nude mouse. An integrin αvβ3 antibody inhibited thyroid hormone-induced cell proliferation in vitro, as did unmodified tetrac and tetrac-NP. Pharmacologic inhibition of the mitogen-activated protein kinase pathway also blocked NSCLC cell proliferation in response to thyroid hormone. Tetrac and tetrac-NP arrested tumor growth and tumor-related angiogenesis in H1299 cells grown in the CAM model and both agents prevented chick embryo mortality. Xenografts of H1299 cells were established in nude mice (n=8, treatment and control groups) and when tumor volumes reached 250-300 mm3, tetrac (1 mg/kg) or tetrac-NP (1mg tetrac as the nanoparticle/kg) were administered intraperitoneally every 2 days. Tetrac and tetrac-NP significantly suppressed tumor growth and angiogenesis. Thus, both tetrac and tetrac-NP effectively arrest human NSCLC tumor cell proliferation in vitro and in the CAM assay and in murine xenograft models.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Tetraiodothyroacetic acid-conjugated PLGA nanoparticles: a nanomedicine approach to treat drug-resistant breast cancer

Dhruba J. Bharali; Murat Yalcin; Paul J. Davis; Shaker A. Mousa

AIM The aim was to evaluate tetraiodothyroacetic acid (tetrac), a thyroid hormone analog of L-thyroxin, conjugated to poly(lactic-co-glycolic acid) nanoparticles (T-PLGA-NPs) both in vitro and in vivo for the treatment of drug-resistant breast cancer. MATERIALS & METHODS The uptake of tetrac and T-PLGA-NPs in doxorubicin-resistant MCF7 (MCF7-Dx) cells was evaluated using confocal microscopy. Cell proliferation assays and a chick chorioallantoic membrane model of FGF2-induced angiogenesis were used to evaluate the anticancer effects of T-PLGA-NPs. In vivo efficacy was examined in a MCF7-Dx orthotopic tumor BALBc nude mouse model. RESULTS T-PLGA-NPs were restricted from entering into the cell nucleus, and T-PLGA-NPs inhibited angiogenesis by 100% compared with 60% by free tetrac. T-PLGA-NPs enhanced inhibition of tumor-cell proliferation at a low-dose equivalent of free tetrac. In vivo treatment with either tetrac or T-PLGA-NPs resulted in a three- to five-fold inhibition of tumor weight. CONCLUSION T-PLGA-NPs have high potential as anticancer agents, with possible applications in the treatment of drug-resistant cancer.


OncoTargets and Therapy | 2014

Nanotetrac targets integrin αvβ3 on tumor cells to disorder cell defense pathways and block angiogenesis

Paul J. Davis; Hung Yun Lin; Thangirala Sudha; Murat Yalcin; Heng Yuan Tang; Aleck Hercbergs; John T. Leith; Mary K. Luidens; Osnat Ashur-Fabian; Sandra Incerpi; Shaker A. Mousa

The extracellular domain of integrin αvβ3 contains a receptor for thyroid hormone and hormone analogs. The integrin is amply expressed by tumor cells and dividing blood vessel cells. The proangiogenic properties of thyroid hormone and the capacity of the hormone to promote cancer cell proliferation are functions regulated nongenomically by the hormone receptor on αvβ3. An L-thyroxine (T4) analog, tetraiodothyroacetic acid (tetrac), blocks binding of T4 and 3,5,3′-triiodo-L-thyronine (T3) by αvβ3 and inhibits angiogenic activity of thyroid hormone. Covalently bound to a 200 nm nanoparticle that limits its activity to the cell exterior, tetrac reformulated as Nanotetrac has additional effects mediated by αvβ3 beyond the inhibition of binding of T4 and T3 to the integrin. These actions of Nanotetrac include disruption of transcription of cell survival pathway genes, promotion of apoptosis by multiple mechanisms, and interruption of repair of double-strand deoxyribonucleic acid breaks caused by irradiation of cells. Among the genes whose expression is suppressed by Nanotetrac are EGFR, VEGFA, multiple cyclins, catenins, and multiple cytokines. Nanotetrac has been effective as a chemotherapeutic agent in preclinical studies of human cancer xenografts. The low concentrations of αvβ3 on the surface of quiescent nonmalignant cells have minimized toxicity of the agent in animal studies.


Bioorganic & Medicinal Chemistry Letters | 2009

Semisynthesis and pharmacological activities of Tetrac analogs: Angiogenesis modulators

Alexandre Bridoux; Huadong Cui; Evgeny Dyskin; Murat Yalcin; Shaker A. Mousa

Novel Tetrac analogs were synthesized and then tested. Anti-angiogenesis efficacy was carried out using the Chick Chorioallantoic Membrane (CAM) model and the mouse matrigel model for angiogenesis. Pharmacological activities showed Tetrac can accommodate numerous modifications and maintain anti-angiogenesis activity.


Cancer Letters | 2014

Suppression of pancreatic cancer by sulfated non-anticoagulant low molecular weight heparin

Thangirala Sudha; Murat Yalcin; Hung Yun Lin; Ahmed M. Elmetwally; Tipu Nazeer; Thiruvengadam Arumugam; Patricia Phillips; Shaker A. Mousa

Sulfated non-anticoagulant heparins (S-NACHs) might be preferred for potential clinical use in cancer patients without affecting hemostasis as compared to low molecular weight heparins (LMWHs). We investigated anti-tumor effects, anti-angiogenesis effects, and mechanisms of S-NACH in a mouse model of pancreatic cancer as compared to the LMWH tinzaparin. S-NACH or tinzaparin with or without gemcitabine were administered, and tumor luminescent signal intensity, tumor weight, and histopathology were assessed at the termination of the study. S-NACH and LMWH efficiently inhibited tumor growth and metastasis, without any observed bleeding events with S-NACH as compared to tinzaparin. S-NACH distinctly increased tumor necrosis and enhanced gemcitabine response in the mouse pancreatic cancer models. These data suggest the potential implication of S-NACH as a neoadjuvant in pancreatic cancer.


Nanomedicine: Nanotechnology, Biology and Medicine | 2017

Targeted delivery of cisplatin to tumor xenografts via the nanoparticle component of nano-diamino-tetrac

Thangirala Sudha; Dhruba J. Bharali; Murat Yalcin; Noureldien H. E. Darwish; Melis Debreli Coskun; Kelly A. Keating; Hung Yun Lin; Paul J. Davis; Shaker A. Mousa

AIM Nano-diamino-tetrac (NDAT) targets a receptor on integrin αvβ3; αvβ3 is generously expressed by cancer cells and dividing endothelial cells and to a small extent by nonmalignant cells. The tetrac (tetraiodothyroacetic acid) of NDAT is covalently bound to a poly(lactic-co-glycolic acid) nanoparticle that encapsulates anticancer drugs. We report NDAT delivery efficiency of cisplatin to agent-susceptible urinary bladder cancer xenografts. MATERIALS & METHODS Cisplatin-loaded NDAT (NDAT-cisplatin) was administered to xenograft-bearing nude mice. Tumor size response and drug content were measured. RESULTS Intratumoral drug concentration was up to fivefold higher (p < 0.001) in NDAT-cisplatin-exposed lesions than with conventional systemic administration. Tumor volume reduction achieved was NDAT-cisplatin > NDAT without cisplatin > cisplatin alone. CONCLUSION NDAT markedly enhances cisplatin delivery to urinary bladder cancer xenografts and increases drug efficacy.


Bioorganic & Medicinal Chemistry Letters | 2010

Semisynthesis and pharmacological activities of thyroxine analogs: Development of new angiogenesis modulators

Alexandre Bridoux; Huadong Cui; Evgeny Dyskin; Andreea-Ruxandra Schmitzer; Murat Yalcin; Shaker A. Mousa

Novel thyroxine analogs with hindered phenol, amino and carboxylic acid groups have been synthesized and the effects of the synthesized compounds on angiogenesis using the chick chorioallantoic membrane and mouse matrigel models have been tested. Pharmacological profiles revealed that thyroxine tolerates numerous modifications on the amino group and remains active. These results provide the rationale for the selection of a novel thyroxine nanoparticle precursor.


Journal of Controlled Release | 2017

Self-assembly of green tea catechin derivatives in nanoparticles for oral lycopene delivery

Weikun Li; Murat Yalcin; Qishan Lin; Mohammed-Salleh M. Ardawi; Shaker A. Mousa

Abstract Lycopene is a natural anti‐oxidant that has attracted much attention due to its varied applications such as protection against loss of bone mass, chronic diseases, skin cancer, prostate cancer, and cardiovascular disease. However, high instability and extremely low oral bioavailability limit its further clinical development. We selected a green tea catechin derivative, oligomerized (‐)‐epigallocatechin‐3‐O‐gallate (OEGCG) as a carrier for oral lycopene delivery. Lycopene‐loaded OEGCG nanoparticles (NPs) were prepared by a nano‐precipitation method, followed by coating with chitosan to form a shell. This method not only can easily control the size of the NP to be around 200 nm to improve its bioavailability, but also can effectively protect the lycopene against degradation due to EGCGs anti‐oxidant property. OEGCG was carefully characterized with nuclear magnetic resonance spectroscopy and mass spectrometry. Lycopene‐loaded polylactic‐co‐glycolic acid (PLGA) NPs were prepared by the same method. Chitosan‐coated OEGCG/lycopene NPs had a diameter of 152 ± 32 nm and a &zgr;‐potential of 58.3 ± 4.2 mv as characterized with transmission electron microscopy and dynamic light scattering. The loading capacity of lycopene was 9% and encapsulation efficiency was 89%. FT‐IR spectral analysis revealed electrostatic interaction between OEGCG and chitosan. Freeze drying of the NPs was also evaluated as a means to improve shelf life. Dynamic light scattering data showed that no aggregation occurred, and the size of the NP increased 1.2 times (Sf / Si ratio) in the presence of 10% sucrose after freeze drying. The in vitro release study showed slow release of lycopene in simulated gastric fluid at acidic pH and faster release in simulated intestinal fluid. In an in vivo study in mice, lycopene pharmacokinetic parameters were improved by lycopene/OEGCG/chitosan NPs, but not improved by lycopene/PLGA/chitosan NPs. The self‐assembled nanostructure of OEGCG combined with lycopene may be a promising application in oral drug delivery in various indications. Graphical abstract Chemical structures of oligomerized (‐)‐epigallocatechin‐3‐O‐gallate (OEGCG) synthesized from the intermolecular polycondensation reaction of EGCG, and a schematic of the self‐assembly process used to form the lycopene/OEGCG/chitosan nanoparticles (NPs), which are formed via two sequential self‐assembly processes in an aqueous solution: complexation of OEGCG with lycopene to form the core, followed by coating with chitosan to form the shell. Figure. No Caption available.

Collaboration


Dive into the Murat Yalcin's collaboration.

Top Co-Authors

Avatar

Shaker A. Mousa

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Evgeny Dyskin

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Thangirala Sudha

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Dhruba J. Bharali

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

Shaymaa S. Mousa

Albany College of Pharmacy and Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge