Network


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

Hotspot


Dive into the research topics where Seetharama D. Satyanarayanajois is active.

Publication


Featured researches published by Seetharama D. Satyanarayanajois.


Journal of Medicinal Chemistry | 2012

Phthalocyanine-Peptide Conjugates for Epidermal Growth Factor Receptor Targeting

Benson G. Ongarora; Krystal R. Fontenot; Xiaoke Hu; Inder Sehgal; Seetharama D. Satyanarayanajois; M. Graça H. Vicente

Four phthalocyanine (Pc)-peptide conjugates designed to target the epidermal growth factor receptor (EGFR) were synthesized and evaluated in vitro using four cell lines: human carcinoma A431 and HEp2, human colorectal HT-29, and kidney Vero (negative control) cells. Two peptide ligands for EGFR were investigated: EGFR-L1 and -L2, bearing 6 and 13 amino acid residues, respectively. The peptides and Pc-conjugates were shown to bind to EGFR using both theoretical (Autodock) and experimental (SPR) investigations. The Pc-EGFR-L1 conjugates 5a and 5b efficiently targeted EGFR and were internalized, in part due to their cationic charge, whereas the uncharged Pc-EGFR-L2 conjugates 4b and 6a poorly targeted EGFR maybe due to their low aqueous solubility. All conjugates were nontoxic (IC(50) > 100 μM) to HT-29 cells, both in the dark and upon light activation (1 J/cm(2)). Intravenous (iv) administration of conjugate 5b into nude mice bearing A431 and HT-29 human tumor xenografts resulted in a near-IR fluorescence signal at ca. 700 nm, 24 h after administration. Our studies show that Pc-EGFR-L1 conjugates are promising near-IR fluorescent contrast agents for CRC and potentially other EGFR overexpressing cancers.


Chemical Biology & Drug Design | 2009

Design, synthesis, and docking studies of peptidomimetics based on HER2-herceptin binding site with potential antiproliferative activity against breast cancer cell lines.

Seetharama D. Satyanarayanajois; Stephanie Villalba; Liu Jianchao; Go Mei Lin

Epidermal growth factor receptor kinase and the related human epidermal growth factor receptor‐2 (HER2, ErbB2) are two growth factor receptors that have implications in cancer. The overexpression or activation of HER2 occurs frequently in breast, ovarian, and lung cancers, making it an important therapeutic target in the treatment of cancer. Blocking HER2‐mediated signaling with antibodies or small molecules has been shown to be effective in inhibiting cell growth. After analyzing the crystal structure of the HER2–herceptin complex, several peptidomimetics (HERP5, 6, and 7) were designed to inhibit HER2‐mediated signaling for cell growth. We have used an in silico screening method to investigate the chemical diversity of the designed compounds. autodock software was used to dock the different analogs of HERP5 and HERP7 with HER2 protein extracellular domain. A total of 53 compounds were docked to HER2 protein, and their binding modes were analyzed in terms of docking energy, hydrogen bonding, and hydrophobic interactions. Compounds that exhibited low‐energy docked structures were chosen for chemical synthesis and biological activity. Two of the compounds (HERP5 and HERP7) exhibited antiproliferative activity, with IC50 values of 0.396 μm and 0.143 μm, respectively, against SKBR‐3 cell lines (breast cancer cell lines) that overexpress HER2 protein.


Journal of Biomolecular Structure & Dynamics | 2012

Inhibition of Protein-Protein Interaction of HER2-EGFR and HER2–HER3 by a Rationally Designed Peptidomimetic

Sashikanth Banappagari; Miriam Corti; Seth H. Pincus; Seetharama D. Satyanarayanajois

Protein–protein interactions (PPI) play a crucial role in many biological processes and modulation of PPI using small molecules to target hot spots has therapeutic value. As a model system we will use PPI of human epidermal growth factor receptors (EGFRs). Among the four EGFRs, EGFR–HER2 and HER2–HER3 are well known in cancer. We have designed a small molecule that is targeted to modulate HER2-mediated signaling. Our approach is novel because the small molecule designed disrupts dimerization not only of EGFR–HER2, but also of HER2–HER3. In the present study we have shown, using surface plasmon resonance analysis, that a peptidomimetic, compound 5, binds specifically to HER2 protein extracellular domain and disrupts the dimerization of EGFRs. To evaluate the effect of compound 5 on HER2 signaling in vitro, Western blot and PathHunter assays were used. Results indicated that compound 5 inhibits the phosphorylation of HER2 kinase domain and inhibits the heterodimerization in a dose-dependent manner. Molecular modeling methods were used to model the PPI of HER2–HER3 heterodimer.


Journal of Medicinal Chemistry | 2011

Conformationally Constrained Peptides from CD2 to Modulate Protein-Protein Interactions between CD2 and CD58

Ameya Gokhale; Thomas K. Weldeghiorghis; Veena Taneja; Seetharama D. Satyanarayanajois

Cell adhesion molecule CD2 and its ligand CD58 provide good examples of protein-protein interactions in cells that participate in the immune response. To modulate the cell adhesion interaction, peptides were designed from the discontinuous epitopes of the β-strand region of CD2 protein. The two strands were linked by a peptide bond. β-Strands in the peptides were nucleated by inserting a β-sheet-inducing (D)-Pro-Pro sequence or a dibenzofuran (DBF) turn mimetic with key amino acid sequences from CD2 protein that binds to CD58. The solution structures of the peptides (5-10) were studied by NMR and molecular dynamics simulations. The ability of these peptides to inhibit cell adhesion interaction was studied by E-rosetting and lymphocyte epithelial assays. Peptides 6 and 7 inhibit the cell adhesion activity with an IC(50) of 7 and 11 nM, respectively, in lymphocyte epithelial adhesion assay. NMR and molecular modeling results indicated that peptides 6 and 7 exhibited β-hairpin structure in solution.


European Journal of Pharmaceutical Sciences | 2013

Concurrent delivery of tocotrienols and simvastatin by lipid nanoemulsions potentiates their antitumor activity against human mammary adenocarcenoma cells

Alaadin Alayoubi; John F. Anderson; Seetharama D. Satyanarayanajois; Paul W. Sylvester; Sami Nazzal

Tocotrienol rich fraction (TRF) of vitamin E was previously shown to have anticancer activity against murine tumor cells in vitro. TRF was also shown to potentiate the anticancer activity of statins. The objectives of this study were therefore (a) to prepare and characterize stable parenteral lipid nanoemulsions as a novel platform for the concurrent delivery of TRF and simvastatin for subsequent use in combination chemotherapy, and (b) to evaluate the antiproliferative activity of the nanoemulsions against MCF-7 and MDA-MB-231 human mammary tumor cells. Nanoemulsions were prepared by the high-pressure homogenization technique using a viscous 70/30 blend of TRF and medium chain triglycerides as the oil phase in which simvastatin was dissolved at 9%w/w loading. Nanoemulsion droplets were about 200 nm in size and had surface potential of -45 mV. In a dissolution study, approximately 20% of simvastatin was released in sink conditions after 24h. The stability of the nanoemulsions was monitored over 6 months of storage. No oxidation or degradation products were detected and no loss in simvastatin loading was observed during this period. The antiproliferative activity of the nanoemulsions was also retained after storage. The IC50 of the TRF nanoemulsions against MCF-7 and MDA-MB-231 was 14 and 7 μM, respectively, which decreased to 10 μM and 4.8 μM when simvastatin was added to the nanoemulsions. Nanoemulsions prepared with tocopherol had no anticancer activity and were used as negative control. This study demonstrated that parenteral lipid nanoemulsions are viable delivery platform for potential use in cancer chemotherapy.


Toxicon | 2010

Venom neutralization by purified bioactive molecules: Synthetic peptide derivatives of the endogenous PLA2 inhibitory protein PIP (a mini-review)

Maung Maung Thwin; Ramar Perumal Samy; Seetharama D. Satyanarayanajois; P. Gopalakrishnakone

Envenomation due to snakebite constitutes a significant public health problem in tropical and subtropical countries. Antivenom therapy is still the mainstay of treatment for snake envenomation, and yet despite recent research focused on the prospects of using antivenom adjuncts to aid in serotherapy, no new products have emerged so far for therapeutic use. Various methodologies including molecular biology, crystallography, functional and morphological approaches, etc., are employed in the search for such inhibitors with a view to generate molecules that can stop partially or completely the activities of toxic phospholipase A(2) (PLA(2)) and snake venom metalloproteinase (SvMPs) enzymes at the molecular level. Herein, both natural and synthetic inhibitors derived from a variety of sources including medicinal plants, mammals, marine animals, fungi, bacteria, and from the venom and blood of snakes have been briefly reviewed. Attention has been focused on the snake serum-based phospholipase A(2) inhibitors (PLIs), particularly on the PLI derived from python snake serum (PIP), highlighting the potential of the natural product, PIP, or possible derivatives of it, as a complementary treatment to serotherapy against the inflammation and/or muscle-damaging activity of snake venoms. The data indicate a more efficient pathway for inhibition and blocking the activity of PLA(2)s and matrix metalloproteinases (MMPs), thus representing a feasible complementary treatment for snakebites. Such information may be helpful for interfering on the biological processes that these molecules are involved in human inflammatory-related diseases, and also for the development of new drugs for treatment of snake envenomation.


Immunotherapy | 2014

Peptides and peptidomimetics as immunomodulators

Ameya Gokhale; Seetharama D. Satyanarayanajois

Peptides and peptidomimetics can function as immunomodulating agents by either blocking the immune response or stimulating the immune response to generate tolerance. Knowledge of B- or T-cell epitopes along with conformational constraints is important in the design of peptide-based immunomodulating agents. Work on the conformational aspects of peptides, synthesis and modified amino acid side chains have contributed to the development of a new generation of therapeutic agents for autoimmune diseases and cancer. The design of peptides/peptidomimetics for immunomodulation in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, systemic lupus and HIV infection is reviewed. In cancer therapy, peptide epitopes are used in such a way that the body is trained to recognize and fight the cancer cells locally as well as systemically.


The International Journal of Biochemistry & Cell Biology | 2012

The opposite effects of doxorubicin on bone marrow stem cells versus breast cancer stem cells depend on glucosylceramide synthase.

Kaustubh N. Bhinge; Vineet Gupta; Salman B. Hosain; Seetharama D. Satyanarayanajois; Sharon A. Meyer; Benny L. Blaylock; Qian-Jin Zhang; Yong-Yu Liu

Myelosuppression and drug resistance are common adverse effects in cancer patients with chemotherapy, and those severely limit the therapeutic efficacy and lead treatment failure. It is unclear by which cellular mechanism anticancer drugs suppress bone marrow, while drug-resistant tumors survive. We report that due to the difference of glucosylceramide synthase (GCS), catalyzing ceramide glycosylation, doxorubicin (Dox) eliminates bone marrow stem cells (BMSCs) and expands breast cancer stem cells (BCSCs). It was found that Dox decreased the numbers of BMSCs (ABCG2(+)) and the sphere formation in a dose-dependent fashion in isolated bone marrow cells. In tumor-bearing mice, Dox treatments (5mg/kg, 6 days) decreased the numbers of BMSCs and white blood cells; conversely, those treatments increased the numbers of BCSCs (CD24(-)/CD44(+)/ESA(+)) more than threefold in the same mice. Furthermore, therapeutic-dose of Dox (1mg/kg/week, 42 days) decreased the numbers of BMSCs while it increased BCSCs in vivo. Breast cancer cells, rather than bone marrow cells, highly expressed GCS, which was induced by Dox and correlated with BCSC pluripotency. These results indicate that Dox may have opposite effects, suppressing BMSCs versus expanding BCSCs, and GCS is one determinant of the differentiated responsiveness of bone marrow and cancer cells.


Future Medicinal Chemistry | 2011

Medicinal chemistry for 2020.

Seetharama D. Satyanarayanajois; Ronald A. Hill

Rapid advances in our collective understanding of biomolecular structure and, in concert, of biochemical systems, coupled with developments in computational methods, have massively impacted the field of medicinal chemistry over the past two decades, with even greater changes appearing on the horizon. In this perspective, we endeavor to profile some of the most prominent determinants of change and speculate as to further evolution that may consequently occur during the next decade. The five main angles to be addressed are: protein-protein interactions; peptides and peptidomimetics; molecular diversity and pharmacological space; molecular pharmacodynamics (significance, potential and challenges); and early-stage clinical efficacy and safety. We then consider, in light of these, the future of medicinal chemistry and the educational preparation that will be required for future medicinal chemists.


MedChemComm | 2011

Structure–activity relationship of conformationally constrained peptidomimetics for antiproliferative activity in HER2-overexpressing breast cancer cell lines

Sashikanth Banappagari; Sharon Ronald; Seetharama D. Satyanarayanajois

Human epidermal growth factor receptor 2 (HER2) is a member of the human epidermal growth factor receptor kinases and is involved in a signaling cascade for cell growth and differentiation. It is well established that HER2-mediated heterodimerization has important implications in cancer. Deregulation of signaling pathways and overexpression of HER2 is known to occur in cancer cells, indicating the role of HER2 in tumorigenesis. Therefore, blocking HER2-mediated signaling has potential therapeutic value. We have designed several peptidomimetics to inhibit HER2-mediated signaling for cell growth. One of the compounds (compound 5, Arg-[3-amino-3(1-napthyl)-propionic acid]-Phe) exhibited antiproliferative activity with IC(50) values in the nanomolar to micromolar range in breast cancer cell lines. To further investigate the structure-activity relationship of the compounds, various analogs of compound 5 were designed. Conformational constraints were initiated in the peptidomimetic with introduction of a Pro residue in the peptidomimetic sequence. Results of antiproliferative activity indicated that analogs of compound 5 with C-and N-terminal ends capped (compound 16) and compound 9 with Asp at the C-terminal exhibited antiproliferative activity in the lower micromolar range against breast cancer cell lines. Introduction of conformational constraints such as Pro residue in the sequence or cyclization did not enhance the activity of the peptidomimetic. Competitive binding studies were carried out to evaluate the binding of potent peptidomimetics to HER2-overexpressing cancer cell lines. Results indicated that compounds exhibiting antiproliferative activity in breast cancer cell lines bind to the cells that overexpress HER2 protein.

Collaboration


Dive into the Seetharama D. Satyanarayanajois's collaboration.

Top Co-Authors

Avatar

Alaadin Alayoubi

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Ameya Gokhale

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Paul W. Sylvester

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Sami Nazzal

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Sashikanth Banappagari

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

P. Gopalakrishnakone

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Jining Liu

Estée Lauder Companies

View shared research outputs
Top Co-Authors

Avatar

John F. Anderson

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge