Network


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

Hotspot


Dive into the research topics where Nisha Kuzhuppilly Ramakrishnan is active.

Publication


Featured researches published by Nisha Kuzhuppilly Ramakrishnan.


Current Pharmaceutical Design | 2010

Sigma Receptors in Oncology: Therapeutic and Diagnostic Applications of Sigma Ligands

Aren van Waarde; Anna A. Rybczynska; Nisha Kuzhuppilly Ramakrishnan; Kiichi Ishiwata; Philip H. Elsinga; Rudi Dierckx

Sigma receptors (subtypes sigma-1 and sigma-2) are a unique class of binding sites expressed throughout the mammalian body. The endogenous ligand for these sites has not been identified, but steroid hormones (particularly progesterone), sphingolipid-derived amines and N,N-dimethyltryptamine can bind with fairly high affinity. Sigma receptors are overexpressed in rapidly proliferating cells, like cancer cells. Particularly the sigma-2 subtype is upregulate when cells divide and down regulated when they become quiescent. Sigma ligands, especially sigma-2 agonists, can inhibit proliferation and induce apoptosis by a mechanism involving changes in cytosolic Ca(2+), ceramide and sphingolipid levels. Tumor cells are much more sensitive to such treatment than cells from their tissue of origin. Sigma ligands induce apoptosis not only in drug-sensitive but also in drug-resistant cancer cells (e.g., cells with p53 mutations, or caspase dysfunction). Moreover, sigma ligands may abrogate P-glycoprotein-mediated drug resistance and at subtoxic doses, they can potentiate the effect of conventional cytostatics. Thus, sigma-2 agonists may be developed as antineoplastic agents for the treatment of drug-resistant tumors. A large number of radiolabeled sigma ligands has been prepared for SPECT (single-photon emission computed tomography) and PET (positron emission tomography) imaging. Such radiopharmaceuticals can be used for tumor detection, tumor staging, and evaluation of anti-tumor therapy. There is still a need for the development of ligands with (1) high selectivity for the sigma-2 subtype, (2) defined action (agonist or antagonist) and (3) optimal pharmacokinetics (low affinity for P-glycoprotein, high and specific tumor uptake, and rapid washout from non-target tissues).


Behavioural Brain Research | 2011

The cholinergic system, sigma-1 receptors and cognition

Aren van Waarde; Nisha Kuzhuppilly Ramakrishnan; Anna A. Rybczynska; Philip H. Elsinga; Kiichi Ishiwata; Ingrid M. Nijholt; Paul G.M. Luiten; Rudi Dierckx

This article provides an overview of present knowledge regarding the relationship between the cholinergic system and sigma-1 receptors, and discusses potential applications of sigma-1 receptor agonists in the treatment of memory deficits and cognitive disorders. Sigma-1 receptors, initially considered as a subtype of the opioid family, are unique ligand-regulated molecular chaperones in the endoplasmatic reticulum playing a modulatory role in intracellular calcium signaling and in the activity of several neurotransmitter systems, particularly the cholinergic and glutamatergic pathways. Several central nervous system (CNS) drugs show high to moderate affinities for sigma-1 receptors, including acetylcholinesterase inhibitors (donepezil), antipsychotics (haloperidol, rimcazole), selective serotonin reuptake inhibitors (fluvoxamine, sertraline) and monoamine oxidase inhibitors (clorgyline). These compounds can influence cognitive functions both via their primary targets and by activating sigma-1 receptors in the CNS. Sigma-1 agonists show powerful anti-amnesic and neuroprotective effects in a large variety of animal models of cognitive dysfunction involving, among others (i) pharmacologic target blockade (with muscarinic or NMDA receptor antagonists or p-chloroamphetamine); (ii) selective lesioning of cholinergic neurons; (iii) CNS administration of β-amyloid peptides; (iv) aging-induced memory loss, both in normal and senescent-accelerated rodents; (v) neurodegeneration induced by toxic compounds (CO, trimethyltin, cocaine), and (vi) prenatal restraint stress.


Biochimica et Biophysica Acta | 2015

Potential applications for sigma receptor ligands in cancer diagnosis and therapy

Aren van Waarde; Anna A. Rybczynska; Nisha Kuzhuppilly Ramakrishnan; Kiichi Ishiwata; Philip H. Elsinga; Rudi A. J. O. Dierckx

Sigma receptors (sigma-1 and sigma-2) represent two independent classes of proteins. Their endogenous ligands may include the hallucinogen N,N-dimethyltryptamine (DMT) and sphingolipid-derived amines which interact with sigma-1 receptors, besides steroid hormones (e.g., progesterone) which bind to both sigma receptor subpopulations. The sigma-1 receptor is a ligand-regulated molecular chaperone with various ion channels and G-protein-coupled membrane receptors as clients. The sigma-2 receptor was identified as the progesterone receptor membrane component 1 (PGRMC1). Although sigma receptors are over-expressed in tumors and up-regulated in rapidly dividing normal tissue, their ligands induce significant cell death only in tumor tissue. Sigma ligands may therefore be used to selectively eradicate tumors. Multiple mechanisms appear to underlie cell killing after administration of sigma ligands, and the signaling pathways are dependent both on the type of ligand and the type of tumor cell. Recent evidence suggests that the sigma-2 receptor is a potential tumor and serum biomarker for human lung cancer and an important target for inhibiting tumor invasion and cancer progression. Current radiochemical efforts are focused on the development of subtype-selective radioligands for positron emission tomography (PET) imaging. Right now, the mostpromising tracers are [18F]fluspidine and [18F]FTC-146 for sigma-1 receptors and [11C]RHM-1 and [18F]ISO-1 for the sigma-2 subtype. Nanoparticles coupled to sigma ligands have shown considerable potential for targeted delivery of antitumor drugs in animal models of cancer, but clinical studies exploring this strategy in cancer patients have not yet been reported. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Journal of Medicinal Chemistry | 2009

Synthesis and Preclinical Evaluation of Novel PET Probes for P-Glycoprotein Function and Expression

Aren van Waarde; Nisha Kuzhuppilly Ramakrishnan; Anna A. Rybczynska; Philip H. Elsinga; Francesco Berardi; Johan R. de Jong; Chantal Kwizera; Roberto Perrone; Mariangela Cantore; Jurgen W. A. Sijbesma; Rudi Dierckx; Nicola Antonio Colabufo

UNLABELLED P-glycoprotein (P-gp) is an ATP-dependent efflux pump protecting the body against xenobiotics. A P-gp substrate (7) and an inhibitor (6) were labeled with (11)C, resulting in potential tracers of P-gp function and expression. METHODS 6 and 7 were labeled using (11)CH(3)I. (11)C-verapamil was prepared as published previously, using (11)C-methyl triflate. MicroPET scans (with arterial sampling) and biodistribution studies were performed in rats pretreated with saline, cyclosporin A (CsA, 50 mg/kg), or cold 6 (15 mg/kg). RESULTS The radiochemical yields of (11)C-6 and (11)C-7 were approximately 30% with a total synthesis time of 45 min. Cerebral distribution volumes (DV) of (11)C-6 (2.35 +/- 0.11) and (11)C-7 (1.86 +/- 0.15) in saline-treated rats were higher than of (11)C-verapamil (0.64 +/- 0.12). DVs of (11)C-7 and (11)C-verapamil were significantly increased by CsA (to 5.26 +/- 0.14 and 5.85 +/- 0.32, respectively). The DV of (11)C-6 was reduced by cold 6 (to 1.65 +/- 0.03). Its uptake was also reduced (up to 67%) in several peripheral organs that express P-gp. CONCLUSIONS (11)C-7 is a novel tracer of P-gp function with higher baseline uptake than (11)C-verapamil. Upregulation of P-gp function in response to treatment (which is hard to detect with (11)C-verapamil) may be detectable using (11)C-7 and PET. Because (11)C-6 shows specific binding in target organs, this compound is the first PET tracer allowing measurement of P-gp expression.


The Journal of Nuclear Medicine | 2013

Small-Animal PET with a σ-Ligand, 11C-SA4503, Detects Spontaneous Pituitary Tumors in Aged Rats

Nisha Kuzhuppilly Ramakrishnan; Anna A. Rybczynska; Anniek K. D. Visser; Krisztina Marosi; Csaba Nyakas; Chantal Kwizera; Jurgen W. A. Sijbesma; Philip H. Elsinga; Kiichi Ishiwata; Jan Pruim; Rudi Dierckx; Aren van Waarde

Pituitary tumors are often detected only after death or at late stages of the disease when they are macroadenomas with a low surgical cure rate. Spontaneous pituitary tumors occur in rats over 1 y of age. In an ongoing study of changes in σ-1 agonist binding related to aging, several of our rats developed such tumors. The aim of the current study was to assess the kinetics of 11C-SA4503 (11C-labeled 1-[2-(3,4-dimethoxyphenthyl)]-4-(3-phenylpropyl)-piperazine dihydrochloride) in tumor and brain and to evaluate the utility of this tracer in the detection of pituitary tumors. Methods: Small-animal PET scans of the brain region of male Wistar Hannover rats (age, 18–32 mo) were acquired using the σ-1 agonist tracer 11C-SA4503. The time-dependent uptake of 11C in the entire brain, tumor or normal pituitary, and thyroid was measured. A 2-tissue-compartment model was fitted to the PET data, using metabolite-corrected plasma radioactivity as the input function. Results: Pituitary tumors showed up as bright hot spots in the scans. The total distribution volume (VT) of the tracer was significantly higher in the tumor than in the normal pituitary. Surprisingly, a higher VT was also seen in the brain and thyroid tissue of animals with pituitary tumors than in healthy rats. The increase in VT in the brain and thyroid was not related to a change in nondisplaceable binding potential (BPND) but rather to an increase in the partition coefficient (K1/k2) of 11C-SA4503. The increase in VT in the tumor on the other hand was accompanied by a significant increase in BPND. Western blotting analysis indicated that pituitary tumors overexpressed σ-1 receptors. Conclusion: The overexpression of σ-1 receptors in spontaneous pituitary tumors is detected as an increase in uptake and BPND of 11C-SA4503. Therefore, this tracer may have promise for the detection of pituitary adenomas, using PET.


European Journal of Nuclear Medicine and Molecular Imaging | 2012

Population pharmacokinetics of cutamesine in rats using NONMEM, C-11-SA4503, and microPET

Nisha Kuzhuppilly Ramakrishnan; V. Pilla Reddy; Johannes H. Proost; Csaba Nyakas; Chantal Kwizera; Jurgen W. A. Sijbesma; Philippus Elsinga; K. Ishiwata; Rudi Dierckx; van Aren Waarde

C. Asferg, R. Møgelvang, A. Flyvbjerg, A. Frystyk, J.S. Jensen, J.L. Marott, M. Appleyard, P. Schnoh, G.B. Jensen, J. Jeppesen. Copenhagen University Hospital Glostrup, Department of Clinical Physiology and Nuclear Medicine, Glostrup, Denmark, Copenhagen University Hospital Gentofte, Department of Cardiology, Gentofte, Denmark, Aarhus University Hospital Aarhus, Medical Research Laboratories, Clinical Institute and Medical Department M, Aarhus, Denmark, Copenhagen University Hospital Bispebjerg, The Copenhagen City Heart Study, Bispebjerg, Denmark, Copenhagen University Hospital Hvidovre, Department of Cardiology, Hvidovre, Denmark, Copenhagen University Hospital Glostrup, Department of Medicine, Glostrup, Denmark, Faculty of Health Sciences, University of Aarhus, Aarhus, Denmark, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark


The Journal of Nuclear Medicine | 2013

In vivo responses of human A375M melanoma to a σ ligand: 18F-FDG PET imaging.

Anna A. Rybczynska; Marco de Bruyn; Nisha Kuzhuppilly Ramakrishnan; Johan R. de Jong; Philip H. Elsinga; Wijnand Helfrich; Rudi Dierckx; Aren van Waarde

σ-ligands can kill tumor cells. Previously we have shown that a short in vitro incubation of C6 tumor cells with σ-ligands (24 h) results in a dose-dependent increase of cellular 18F-FDG uptake and that the magnitude of this increase is predictive of subsequent cell death. Here, we aimed to assess whether the σ-ligand rimcazole inhibits growth of A375M melanoma xenografts in nude mice and whether rimcazole treatment changes 18F-FDG uptake in vivo. Methods: Athymic mice were inoculated with A375M melanoma cells. After 2 wk, tumors had reached a size of 41 ± 6 mm3. We then started a 14-d treatment schedule with daily drug dosing. Control animals were injected with water and treated animals with rimcazole (26 mg/kg) in water. Three small-animal PET scans with 18F-FDG were obtained: on days 0, 7, and 14 of treatment. After the last scan, animals were terminated, and a biodistribution study was performed. Results: Rimcazole treatment resulted in a greater than 4-fold reduction of tumor weight in comparison to controls at day 14 (100 ± 26 vs. 436 ± 117 mg, respectively, P < 0.03). Treatment did not affect the levels of (nonradioactive) glucose in blood, σ-1 and σ-2 receptor expression in the tumor, animal weight, behavior, or appearance. Antitumor activity of rimcazole was accompanied by a transient increase of the tumor uptake of 18F-FDG (measured at day 7). Significant increases of 18F-FDG uptake at day 14 were observed in the liver and pancreas. Conclusion: Rimcazole strongly inhibited the growth of A375M melanoma xenografts. This growth inhibition is accompanied by an early increase of 18F-FDG uptake in the tumor.


Journal of Cerebral Blood Flow and Metabolism | 2014

[(11)C]5-HTP and microPET are not suitable for pharmacodynamic studies in the rodent brain.

Anniek K. D. Visser; Nisha Kuzhuppilly Ramakrishnan; Antoon T. M. Willemsen; Valentina Di Gialleonardo; Erik F. J. de Vries; Ido P. Kema; Rudi A. J. O. Dierckx; Aren van Waarde

The PET tracer [11C]5-hydroxytryptophan ([11C]5-HTP), which is converted to [11C]5-hydroxytryptamine ([11C]5-HT) by aromatic amino acid decarboxylase (AADC), is thought to measure 5-HT synthesis rates. But can we measure these synthesis rates by kinetic modeling of [11C]5-HTP in rat? Male rats were scanned with [11C]5-HTP (60 minutes) after different treatments. Scans included arterial blood sampling and metabolite analysis. 5-HT synthesis rates were calculated by a two-tissue compartment model (2TCM) with irreversible tracer trapping or Patlak analysis. Carbidopa (inhibitor peripheral AADC) dose-dependently increased [11C]5-HTP brain uptake, but did not influence 2TCM parameters. Therefore, 10 mg/kg carbidopa was applied in all subsequent study groups. These groups included treatment with NSD 1015 (general AADC inhibitor) or p-chlorophenylalanine (PCPA, inhibitor of tryptophan hydroxylase, TPH). In addition, the effect of a low-tryptophan (Trp) diet was investigated. NSD 1015 or Trp depletion did not affect any model parameters, but PCPA reduced [11C]5-HTP uptake, and the k3. This was unexpected as NSD 1015 directly inhibits the enzyme converting [11C]5-HTP to [11C]5-HT, suggesting that trapping of radioactivity does not distinguish between parent tracer and its metabolites. As different results have been acquired in monkeys and humans, [11C]5-HTP-PET may be suitable for measuring 5-HT synthesis in primates, but not in rodents.


Psychopharmacology | 2014

Dose-dependent sigma-1 receptor occupancy by donepezil in rat brain can be assessed with C-11-SA4503 and microPET (vol 231, pg 3997, 2014)

Nisha Kuzhuppilly Ramakrishnan; Anniek K. D. Visser; Marianne Schepers; Gert Luurtsema; Csaba Nyakas; Philip H. Elsinga; Kiichi Ishiwata; Rudi A. J. O. Dierckx; Aren van Waarde

The original version of this article inadvertently contained a mistake. Figure 4 was corrupted in the original publication of this paper. Here is a corrected and updated version of this figure.


Psychopharmacology | 2015

Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses

Kristoffer Sahlholm; Jurgen W. A. Sijbesma; Bram Maas; Chantal Kwizera; Daniel Marcellino; Nisha Kuzhuppilly Ramakrishnan; Rudi A. J. O. Dierckx; Philip H. Elsinga; Aren van Waarde

Collaboration


Dive into the Nisha Kuzhuppilly Ramakrishnan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rudi A. J. O. Dierckx

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Chantal Kwizera

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Rudi Dierckx

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna A. Rybczynska

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Anniek K. D. Visser

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Kiichi Ishiwata

Fukushima Medical University

View shared research outputs
Top Co-Authors

Avatar

Jurgen W. A. Sijbesma

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge