Bio


The primary goal of my research is to discover imaging and therapeutic solutions to human diseases. I apply chemistry, radiology, and nanotechnology techniques (for example, organic synthesis, radiochemistry, and bioconjugation with metal and non-metal nanoparticles) to image innate and adaptive immune cells in the brain in the context of neurodegenerative diseases (Stanford), bacterial infections (UCSF), treat human glioma (UCSF), and image disease biomarkers (Utah). My research in the chemistry-nanoscience-glycobiology interface has produced several impactful peer-reviewed publications: 1) a nanosensor that diagnoses life-threatening contaminants in pharmaceutical-grade heparin, an anticoagulant used extensively during surgery, 2) heparan sulfate code readers, 3) sugar PET tracers to image bacterial metabolism. I am currently working towards the development of new neuro-PET tracers at Stanford Medical School. In the future, I seek to combine my organic chemistry, radiosynthesis, and cell biology skills to build an independent research program to develop theranostic solutions (diagnosis and therapeutic) to human diseases.
Specialties: Organic synthesis, Radiochemistry (18F, 11C, 89Zr, 64Cu), material chemistry, carbohydrate chemistry, biochemistry, imaging, neuroimmunology, oncology

Current Role at Stanford


Senior Research Scientist: a) cold chemical synthesis— Synthesis of the 12C and 19F- HPLC standards and precursors for 11C- and 18F- labeling
b) Radiosynthesis— Introduction of 11C or 18F radioisotopes into small molecules to develop novel PET tracers, that can track activated myeloid cells in neurodegenerative disease, c) radiometal labeling— 64Cu and 89Zr labeling of monoclonal antibodies that target immune receptors, d) clinical translation— To follow FDA guidelines for translating preclinically validated tracers into humans in the cyclotron and radiochemistry facility (CRF) of the Stanford University

Honors & Awards


  • Co-investigator, Wu Tsai Translate Award, Wu Tsai Neuroscience Institute, Stanford University (January, 2024)
  • Cover Article (JACS-Au, 2023, 3, 12, 3297-3310) https://pubs.acs.org/doi/10.1021/jacsau.3c00435, American Chemical Society Journal (December, 2023)
  • SNMMI highlights and interview https://www.youtube.com/watch?v=wswYdHf46V0, Society of Nuclear Medicine and Molecular Imaging (June, 2023)
  • Top Abstract, World Molecular Imaging Conference, Montreal (September, 2019)
  • Invited Speaker, Breaking News Session, Gordon Conference- Proteoglycans (July, 2014)
  • Fateley-Hammaker Collaboration in Research Award, Kansas State University (April, 2010)
  • Terry C. Johnson basic cancer research award, Kansas State University (May, 2008)

Professional Affiliations and Activities


  • Senior Scientist, Stanford University (2021 - Present)
  • Assistant Research Professional, University of California— San Francisco (2016 - 2021)
  • Postdoctoral Scholar, University of Utah (2012 - 2016)

All Publications


  • PET Imaging of Innate Immune Activation Using 11C Radiotracers Targeting GPR84. JACS Au Kalita, M., Park, J. H., Kuo, R. C., Hayee, S., Marsango, S., Straniero, V., Alam, I. S., Rivera-Rodriguez, A., Pandrala, M., Carlson, M. L., Reyes, S. T., Jackson, I. M., Suigo, L., Luo, A., Nagy, S. C., Valoti, E., Milligan, G., Habte, F., Shen, B., James, M. L. 2023; 3 (12): 3297-3310

    Abstract

    Chronic innate immune activation is a key hallmark of many neurological diseases and is known to result in the upregulation of GPR84 in myeloid cells (macrophages, microglia, and monocytes). As such, GPR84 can potentially serve as a sensor of proinflammatory innate immune responses. To assess the utility of GPR84 as an imaging biomarker, we synthesized 11C-MGX-10S and 11C-MGX-11Svia carbon-11 alkylation for use as positron emission tomography (PET) tracers targeting this receptor. In vitro experiments demonstrated significantly higher binding of both radiotracers to hGPR84-HEK293 cells than that of parental control HEK293 cells. Co-incubation with the GPR84 antagonist GLPG1205 reduced the binding of both radiotracers by >90%, demonstrating their high specificity for GPR84 in vitro. In vivo assessment of each radiotracer via PET imaging of healthy mice illustrated the superior brain uptake and pharmacokinetics of 11C-MGX-10S compared to 11C-MGX-11S. Subsequent use of 11C-MGX-10S to image a well-established mouse model of systemic and neuro-inflammation revealed a high PET signal in affected tissues, including the brain, liver, lung, and spleen. In vivo specificity of 11C-MGX-10S for GPR84 was confirmed by the administration of GLPG1205 followed by radiotracer injection. When compared with 11C-DPA-713-an existing radiotracer used to image innate immune activation in clinical research studies-11C-MGX-10S has multiple advantages, including its higher binding signal in inflamed tissues in the CNS and periphery and low background signal in healthy saline-treated subjects. The pronounced uptake of 11C-MGX-10S during inflammation, its high specificity for GPR84, and suitable pharmacokinetics strongly support further investigation of 11C-MGX-10S for imaging GPR84-positive myeloid cells associated with innate immune activation in animal models of inflammatory diseases and human neuropathology.

    View details for DOI 10.1021/jacsau.3c00435

    View details for PubMedID 38155640

    View details for PubMedCentralID PMC10751761

  • Application of Machine Learning Driven Computational Approaches for Novel CNS PET Tracer Development Jackson, I., Luo, A., Webb, E., Zhang, B., Guo, A., Nagy, S., Shao, X., Kuo, R., Carlson, M., Alam, I., Rodriguez, A., Winton, W., Stauff, J., Kalita, M., Scott, P., James, M. ELSEVIER SCIENCE INC. 2023: S40-S41
  • Clinical Radiosynthesis and Translation of [18F]OP-801: A Novel Radiotracer for Imaging Reactive Microglia and Macrophages. ACS chemical neuroscience Jackson, I. M., Carlson, M. L., Beinat, C., Malik, N., Kalita, M., Reyes, S., Azevedo, E. C., Nagy, S. C., Alam, I. S., Sharma, R., La Rosa, S. A., Moradi, F., Cleland, J., Shen, B., James, M. L. 2023

    Abstract

    Positron emission tomography (PET) is a powerful tool for studying neuroinflammatory diseases; however, current PET biomarkers of neuroinflammation possess significant limitations. We recently reported a promising dendrimer PET tracer ([18F]OP-801), which is selectively taken up by reactive microglia and macrophages. Here, we describe further important characterization of [18F]OP-801 in addition to optimization and validation of a two-step clinical radiosynthesis. [18F]OP-801 was found to be stable in human plasma for 90 min post incubation, and human dose estimates were calculated for 24 organs of interest; kidneys and urinary bladder wall without bladder voiding were identified as receiving the highest absorbed dose. Following optimization detailed herein, automated radiosynthesis and quality control (QC) analyses of [18F]OP-801 were performed in triplicate in suitable radiochemical yield (6.89 ± 2.23% decay corrected), specific activity (37.49 ± 15.49 GBq/mg), and radiochemical purity for clinical imaging. Importantly, imaging mice with tracer (prepared using optimized methods) 24 h following the intraperitoneal injection of liposaccharide resulted in the robust brain PET signal. Cumulatively, these data enable clinical translation of [18F]OP-801 for imaging reactive microglia and macrophages in humans. Data from three validation runs of the clinical manufacturing and QC were submitted to the Food and Drug Administration (FDA) as part of a Drug Master File (DMF). Subsequent FDA approval to proceed was obtained, and a phase 1/2 clinical trial (NCT05395624) for first-in-human imaging in healthy controls and patients with amyotrophic lateral sclerosis is underway.

    View details for DOI 10.1021/acschemneuro.3c00028

    View details for PubMedID 37310119

  • Development and initial evaluation of a novel 11C-labeled PET tracer to image GPR84 expressing-myeloid cells during neuroinflammation Kalita, M., Park, J., Hayee, S., Marsango, S., Carlson, M., Reyes, S., Nagy, S., Straniero, V., Pandrala, M., Jackson, I., Alam, I., Valoti, E., Milligan, G., Shen, B., James, M. SOC NUCLEAR MEDICINE INC. 2023
  • Antigen-Dependent Inducible T-Cell Reporter System for PET Imaging of Breast Cancer and Glioblastoma. Journal of nuclear medicine : official publication, Society of Nuclear Medicine Shin, J., Parker, M. F., Zhu, I., Alanizi, A., Rodriguez, C. I., Liu, R., Watchmaker, P. B., Kalita, M., Blecha, J., Luu, J., Wright, B., Lapi, S. E., Flavell, R. R., Okada, H., Tlsty, T. D., Roybal, K. T., Wilson, D. M. 2023; 64 (1): 137-144

    Abstract

    For the past several decades, chimeric antigen receptor T-cell therapies have shown promise in the treatment of cancers. These treatments would greatly benefit from companion imaging biomarkers to follow the trafficking of T cells invivo. Methods: Using synthetic biology, we engineered T cells with a chimeric receptor synthetic intramembrane proteolysis receptor (SNIPR) that induces overexpression of an exogenous reporter gene cassette on recognition of specific tumor markers. We then applied a SNIPR-based PET reporter system to 2 cancer-relevant antigens, human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor variant III (EGFRvIII), commonly expressed in breast and glial tumors, respectively. Results: Antigen-specific reporter induction of the SNIPR PET T cells was confirmed invitro using green fluorescent protein fluorescence, luciferase luminescence, and the HSV-TK PET reporter with 9-(4-18F-fluoro-3-[hydroxymethyl]butyl)guanine ([18F]FHBG). T cells associated with their target antigens were successfully imaged using PET in dual-xenograft HER2+/HER2- and EGFRvIII+/EGFRvIII- animal models, with more than 10-fold higher [18F]FHBG signals seen in antigen-expressing tumors versus the corresponding controls. Conclusion: The main innovation found in this work was PET detection of T cells via specific antigen-induced signals, in contrast to reporter systems relying on constitutive gene expression.

    View details for DOI 10.2967/jnumed.122.264284

    View details for PubMedID 35981900

  • Iron-Based Magnetic Nanosystems for Diagnostic Imaging and Drug Delivery: Towards Transformative Biomedical Applications PHARMACEUTICS Bossmann, S. H., Payne, M. M., Kalita, M., Bristow, R. D., Afshar, A., Perera, A. S. 2022; 14 (10)

    Abstract

    The advancement of biomedicine in a socioeconomically sustainable manner while achieving efficient patient-care is imperative to the health and well-being of society. Magnetic systems consisting of iron based nanosized components have gained prominence among researchers in a multitude of biomedical applications. This review focuses on recent trends in the areas of diagnostic imaging and drug delivery that have benefited from iron-incorporated nanosystems, especially in cancer treatment, diagnosis and wound care applications. Discussion on imaging will emphasise on developments in MRI technology and hyperthermia based diagnosis, while advanced material synthesis and targeted, triggered transport will be the focus for drug delivery. Insights onto the challenges in transforming these technologies into day-to-day applications will also be explored with perceptions onto potential for patient-centred healthcare.

    View details for DOI 10.3390/pharmaceutics14102093

    View details for Web of Science ID 000874342700001

    View details for PubMedID 36297529

    View details for PubMedCentralID PMC9607318

  • Glyco-nanotechnology: A biomedical perspective NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE Kalita, M., Payne, M. M., Bossmann, S. H. 2022; 42: 102542

    Abstract

    Glycans govern cellular signaling through glycan-protein and glycan-glycan crosstalk. Disruption in the crosstalk initiates 'rogue' signaling and pathology. Nanomaterials supply platforms for multivalent displays of glycans, mediate 'rogue' signal correction, and provide disease treatment modalities (therapeutics). The decorated glycans also target overexpressed lectins on unhealthy cells and direct metal nanoparticles such as gold, iron oxide, and quantum dots to the site of infection. The nanoparticles inform us about the state of the disease (diagnosis) through their distinct optical, magnetic, and electronic properties. Glyco-nanoparticles can sense disease biomarkers, report changes in protein-glycan interactions, and safeguard quality control (analysis). Here we review the current state of glyco-nanotechnology focusing on diagnosis, therapeutics, and analysis of human diseases. We highlight how glyco-nanotechnology could aid in improving diagnostic methods for the detection of disease biomarkers with magnetic resonance imaging (MRI) and fluorescence imaging (FLI), enhance therapeutics such as anti-adhesive treatment of cancer and vaccines against pneumonia, and advance analysis such as the rapid detection of pharmaceutical heparin contaminant and recombinant SARS-COV-2 spike protein. We illustrate these progressions and outline future potentials of glyco-nanotechnology in advancing human health.

    View details for DOI 10.1016/j.nano.2022.102542

    View details for Web of Science ID 000793782400009

    View details for PubMedID 35189393

  • Radiosynthesis and initial preclinical evaluation of [11C]AZD1283 as a potential P2Y12R PET radiotracer. Nuclear medicine and biology Jackson, I. M., Buccino, P. J., Azevedo, E. C., Carlson, M. L., Luo, A. S., Deal, E. M., Kalita, M., Reyes, S. T., Shao, X., Beinat, C., Nagy, S. C., Chaney, A. M., Anders, D. A., Scott, P. J., Smith, M., Shen, B., James, M. L. 2022

    Abstract

    INTRO: Chronic neuroinflammation and microglial dysfunction are key features of many neurological diseases, including Alzheimer's Disease and multiple sclerosis. While there is unfortunately a dearth of highly selective molecular imaging biomarkers/probes for studying microglia in vivo, P2Y12R has emerged as an attractive candidate PET biomarker being explored for this purpose. Importantly, P2Y12R is selectively expressed on microglia in the CNS and undergoes dynamic changes in expression according to inflammatory context (e.g., toxic versus beneficial/healing states), thus having the potential to reveal functional information about microglia in living subjects. Herein, we identified a high affinity, small molecule P2Y12R antagonist (AZD1283) to radiolabel and assess as a candidate radiotracer through in vitro assays and in vivo positron emission tomography (PET) imaging of both wild-type and total knockout mice and a non-human primate.METHODS: First, we evaluated the metabolic stability and passive permeability of non-radioactive AZD1283 in vitro. Next, we radiolabeled [11C]AZD1283 with radioactive precursor [11C]NH4CN and determined stability in formulation and human plasma. Finally, we investigated the in vivo stability and kinetics of [11C]AZD1283 via dynamic PET imaging of naive wild-type mice, P2Y12R knockout mouse, and a rhesus macaque.RESULTS: We determined the half-life of AZD1283 in mouse and human liver microsomes to be 37 and>160min, respectively, and predicted passive CNS uptake with a small amount of active efflux, using a Caco-2 assay. Our radiolabeling efforts afforded [11C]AZD1283 in an activity of 12.69±10.64mCi with high chemical and radiochemical purity (>99%) and molar activity of 1142.84±504.73mCi/mumol (average of n=3). Of note, we found [11C]AZD1283 to be highly stable in vitro, with >99% intact tracer present after 90min of incubation in formulation and 60min of incubation in human serum. PET imaging revealed negligible brain signal in healthy wild-type mice (n=3) and a P2Y12 knockout mouse (0.55±0.37%ID/g at 5min post injection). Strikingly, high signal was detected in the liver of all mice within the first 20min of administration (peak uptake=58.28±18.75%ID/g at 5min post injection) and persisted for the remaining duration of the scan. Ex vivo gamma counting of mouse tissues at 60min post-injection mirrored in vivo data with a mean %ID/g of 0.9%±0.40, 0.02%±0.01, and 106±29.70% in the blood, brain, and liver, respectively (n=4). High performance liquid chromatography (HPLC) analysis of murine blood and liver metabolite samples revealed a single radioactive peak (relative area under peak: 100%), representing intact tracer. Finally, PET imaging of a rhesus macaque also revealed negligible CNS uptake/binding in monkey brain (peak uptake=0.37 Standard Uptake Values (SUV)).CONCLUSION: Despite our initial encouraging liver microsome and Caco-2 monolayer data, in addition to the observed high stability of [11C]AZD1283 in formulation and human serum, in vivo brain uptake was negligible and rapid accumulation was observed in the liver of both naive wildtype and P2Y12R knockout mice. Liver signal appeared to be independent of both metabolism and P2Y12R expression due to the confirmation of intact tracer in this tissue for both wildtype and P2Y12R knockout mice. In Rhesus Macaque, negligible uptake of [11C]AZD1283 brain indicates a lack of potential for translation or its further investigation in vivo. P2Y12R is an extremely promising potential PET biomarker, and the data presented here suggests encouraging metabolic stability for this scaffold; however, the mechanism of liver uptake in mice should be elucidated prior to further analogue development.

    View details for DOI 10.1016/j.nucmedbio.2022.05.001

    View details for PubMedID 35680502

  • Synthesis and Screening of a-Xylosides in Human Glioblastoma Cells MOLECULAR PHARMACEUTICS Kalita, M., Villanueva-Meyer, J., Ohkawa, Y., Kalyanaraman, C., Chen, K., Mohamed, E., Parker, M. L., Jacobson, M. P., Phillips, J. J., Evans, M. J., Wilson, D. M. 2021; 18 (1): 451-460

    Abstract

    Glycosaminoglycans (GAGs) such as heparan sulfate and chondroitin sulfate decorate all mammalian cell surfaces. These mucopolysaccharides act as coreceptors for extracellular ligands, regulating cell signaling, growth, proliferation, and adhesion. In glioblastoma, the most common type of primary malignant brain tumor, dysregulated GAG biosynthesis results in altered chain length, sulfation patterns, and the ratio of contributing monosaccharides. These events contribute to the loss of normal cellular function, initiating and sustaining malignant growth. Disruption of the aberrant cell surface GAGs with small molecule inhibitors of GAG biosynthetic enzymes is a potential therapeutic approach to blocking the rogue signaling and proliferation in glioma, including glioblastoma. Previously, 4-azido-xylose-α-UDP sugar inhibited both xylosyltransferase (XYLT-1) and β-1,4-galactosyltransferase-7 (β-GALT-7)-the first and second enzymes of GAG biosynthesis-when microinjected into a cell. In another study, 4-deoxy-4-fluoro-β-xylosides inhibited β-GALT-7 at 1 mM concentration in vitro. In this work, we seek to solve the enduring problem of drug delivery to human glioma cells at low concentrations. We developed a library of hydrophobic, presumed prodrugs 4-deoxy-4-fluoro-2,3-dibenzoyl-(α- or β-) xylosides and their corresponding hydrophilic inhibitors of XYLT-1 and β-GALT-7 enzymes. The prodrugs were designed to be activatable by carboxylesterase enzymes overexpressed in glioblastoma. Using a colorimetric MTT assay in human glioblastoma cell lines, we identified a prodrug-drug pair (4-nitrophenyl-α-xylosides) as lead drug candidates. The candidates arrest U251 cell growth at an IC50 = 380 nM (prodrug), 122 μM (drug), and U87 cells at IC50 = 10.57 μM (prodrug). Molecular docking studies were consistent with preferred binding of the α- versus β-nitro xyloside conformer to XYLT-1 and β-GALT-7 enzymes.

    View details for DOI 10.1021/acs.molpharmaceut.0c00839

    View details for Web of Science ID 000606803900036

    View details for PubMedID 33315406

    View details for PubMedCentralID PMC8483608

  • Visualizing antithrombin-binding 3-O-sulfated heparan sulfate motifs on cell surfaces CHEMICAL COMMUNICATIONS Kalita, M., Chua, J., Boothello, R. S., Joice, A., Antelope, O., Roy, A., Babu, P., Saijoh, Y., Desai, U. R., Kuberan, B. 2020; 56 (92): 14423-14426

    Abstract

    To map the cellular topography of the rare 3-O-sulfated structural motif of heparan sulfate (HS), we constructed quantum dot-based probes for antithrombin and FGF2, which reveal widely different distribution of the targeted HS motifs. The technology helps show that old and young aortic endothelia display widely different levels of the antithrombin-binding 3-O-sulfated HS motif.

    View details for DOI 10.1039/d0cc05893a

    View details for Web of Science ID 000591568400018

    View details for PubMedID 33146178

  • Arabinofuranose-derived positron-emission tomography radiotracers for detection of pathogenic microorganisms JOURNAL OF LABELLED COMPOUNDS & RADIOPHARMACEUTICALS Kalita, M., Parker, M. L., Luu, J. M., Stewart, M. N., Blecha, J. E., VanBrocklin, H. F., Evans, M. J., Flavell, R. R., Rosenberg, O. S., Ohliger, M. A., Wilson, D. M. 2020; 63 (5): 231-239

    Abstract

    Detection of bacteria-specific metabolism via positron emission tomography (PET) is an emerging strategy to image human pathogens, with dramatic implications for clinical practice. In silico and in vitro screening tools have recently been applied to this problem, with several monosaccharides including l-arabinose showing rapid accumulation in Escherichia coli and other organisms. Our goal for this study was to evaluate several synthetically viable arabinofuranose-derived 18 F analogs for their incorporation into pathogenic bacteria.We synthesized four radiolabeled arabinofuranose-derived sugars: 2-deoxy-2-[18 F]fluoro-arabinofuranoses (d-2-18 F-AF and l-2-18 F-AF) and 5-deoxy-5-[18 F]fluoro-arabinofuranoses (d-5-18 F-AF and l-5-18 F-AF). The arabinofuranoses were synthesized from 18 F- via triflated, peracetylated precursors analogous to the most common radiosynthesis of 2-deoxy-2-[18 F]fluoro-d-glucose ([18 F]FDG). These radiotracers were screened for their uptake into E. coli and Staphylococcus aureus. Subsequently, the sensitivity of d-2-18 F-AF and l-2-18 F-AF to key human pathogens was investigated in vitro.All 18 F radiotracer targets were synthesized in high radiochemical purity. In the screening study, d-2-18 F-AF and l-2-18 F-AF showed greater accumulation in E. coli than in S. aureus. When evaluated in a panel of pathologic microorganisms, both d-2-18 F-AF and l-2-18 F-AF demonstrated sensitivity to most gram-positive and gram-negative bacteria.Arabinofuranose-derived 18 F PET radiotracers can be synthesized with high radiochemical purity. Our study showed absence of bacterial accumulation for 5-substitued analogs, a finding that may have mechanistic implications for related tracers. Both d-2-18 F-AF and l-2-18 F-AF showed sensitivity to most gram-negative and gram-positive organisms. Future in vivo studies will evaluate the diagnostic accuracy of these radiotracers in animal models of infection.

    View details for DOI 10.1002/jlcr.3835

    View details for Web of Science ID 000521849000001

    View details for PubMedID 32222086

    View details for PubMedCentralID PMC7364301

  • A glycan-based approach to therapeutic angiogenesis PLOS ONE Chua, J., Tran, V. M., Kalita, M., Quintero, M. V., Antelope, O., Muruganandam, G., Saijoh, Y., Kuberan, B. 2017; 12 (8): e0182301

    Abstract

    Angiogenesis, the sprouting of new blood vessels from existing vasculature, involves multiple complex biological processes, and it is an essential step for hemostasis, tissue healing and regeneration. Angiogenesis stimulants can ameliorate human disease conditions including limb ischemia, chronic wounds, heart disease, and stroke. The current strategies to improve the bioavailability of pro-angiogenic growth factors, including VEGF and FGF2, have remained largely unsuccessful. This study demonstrates that small molecules, termed click-xylosides, can promote angiogenesis in the in vitro matrigel tube formation assay and the ex ovo chick chorioallantoic membrane assay, depending on their aglycone moieties. Xyloside treatment enhances network connectivity and cell survivability, thereby, maintaining the network structures on matrigel culture for an extended period of time. These effects were achieved via the secreted xyloside-primed glycosaminoglycans (GAG) chains that in part, act through an ERK1/2 mediated signaling pathway. Through the remodeling of GAGs in the extracellular matrix of endothelial cells, the glycan approach, involving xylosides, offers great potential to effectively promote therapeutic angiogenesis.

    View details for DOI 10.1371/journal.pone.0182301

    View details for Web of Science ID 000406766500059

    View details for PubMedID 28763512

    View details for PubMedCentralID PMC5538652

  • BODIPY-Conjugated Xyloside Primes Fluorescent Glycosaminoglycans in the Inner Ear of Opsanus tau JARO-JOURNAL OF THE ASSOCIATION FOR RESEARCH IN OTOLARYNGOLOGY Holman, H. A., Tran, V. M., Kalita, M., Nguyen, L. N., Arungundram, S., Kuberan, B., Rabbitt, R. D. 2016; 17 (6): 525-540

    Abstract

    We report on a new xyloside conjugated to BODIPY, BX and its utility to prime fluorescent glycosaminoglycans (BX-GAGs) within the inner ear in vivo. When BX is administered directly into the endolymphatic space of the oyster toadfish (Opsanus tau) inner ear, fluorescent BX-GAGs are primed and become visible in the sensory epithelia of the semicircular canals, utricle, and saccule. Confocal and 2-photon microscopy of vestibular organs fixed 4 h following BX treatment, reveal BX-GAGs constituting glycocalyces that envelop hair cell kinocilium, nerve fibers, and capillaries. In the presence of GAG-specific enzymes, the BX-GAG signals are diminished, suggesting that chondroitin sulfates are the primary GAGs primed by BX. Results are consistent with similar click-xylosides in CHO cell lines, where the xyloside enters the Golgi and preferentially initiates chondroitin sulfate B production. Introduction of BX produces a temporary block of hair cell mechanoelectrical transduction (MET) currents in the crista, reduction in background discharge rate of afferent neurons, and a reduction in sensitivity to physiological stimulation. A six-degree-of-freedom pharmacokinetic mathematical model has been applied to interpret the time course and spatial distribution of BX and BX-GAGs. Results demonstrate a new optical approach to study GAG biology in the inner ear, for tracking synthesis and localization in real time.

    View details for DOI 10.1007/s10162-016-0585-5

    View details for Web of Science ID 000388110200002

    View details for PubMedID 27619213

    View details for PubMedCentralID PMC5112219

  • Synthesis and Biomedical Applications of Xylosides GLYCOSAMINOGLYCANS: CHEMISTRY AND BIOLOGY Kalita, M., Quintero, M. V., Raman, K., Tran, V. M., Kuberan, B., Balagurunathan, K., Nakato, H., Desai, U. R. 2015; 1229: 517-528

    Abstract

    Xylosides modulate the biosynthesis of sulfated glycosaminoglycans (GAGs) in various cell types. A new class of xylosides called "click-xylosides" has been synthesized for their biostability, ease of chemical synthesis, and tunable sulfated GAG biogenesis in vitro and in vivo. These click-xylosides have several therapeutic and biomedical applications in the regulation of angiogenesis, tumor inhibition, and regeneration. This protocol focuses on the synthesis of click-xylosides, their cellular priming activities, and biomedical applications.

    View details for DOI 10.1007/978-1-4939-1714-3_40

    View details for Web of Science ID 000344016200041

    View details for PubMedID 25325977

  • A Nanosensor for Ultrasensitive Detection of Oversulfated Chondroitin Sulfate Contaminant in Heparin JOURNAL OF THE AMERICAN CHEMICAL SOCIETY Kalita, M., Balivada, S., Swarup, V., Mencio, C., Raman, K., Desai, U. R., Troyer, D., Kuberan, B. 2014; 136 (2): 554-557

    Abstract

    Heparin has been extensively used as an anticoagulant for the last eight decades. Recently, the administration of a contaminated batch of heparin caused 149 deaths in several countries including USA, Germany, and Japan. The contaminant responsible for the adverse effects was identified as oversulfated chondroitin sulfate (OSCS). Here, we report a rapid, ultrasensitive method of detecting OSCS in heparin using a nanometal surface energy transfer (NSET) based gold-heparin-dye nanosensor. The sensor is an excellent substrate for heparitinase enzyme, as evidenced by ~70% recovery of fluorescence from the dye upon heparitinase treatment. However, the presence of OSCS results in diminished fluorescence recovery from the nanosensor upon heparitinase treatment, as the enzyme is inhibited by the contaminant. The newly designed nanosensor can detect as low as 1 × 10(-9) % (w/w) OSCS making it the most sensitive tool to date for the detection of trace amounts of OSCS in pharmaceutical heparins.

    View details for DOI 10.1021/ja409170z

    View details for Web of Science ID 000330018600004

    View details for PubMedID 24127748

  • Nanoplatforms for highly sensitive fluorescence detection of cancer-related proteases PHOTOCHEMICAL & PHOTOBIOLOGICAL SCIENCES Wang, H., Udukala, D. N., Samarakoon, T. N., Basel, M. T., Kalita, M., Abayaweera, G., Manawadu, H., Malalasekera, A., Robinson, C., Villanueva, D., Maynez, P., Bossmann, L., Riedy, E., Barriga, J., Wang, N., Li, P., Higgins, D. A., Zhu, G., Troyer, D. L., Bossmann, S. H. 2014; 13 (2): 231-240

    Abstract

    Numerous proteases are known to be necessary for cancer development and progression including matrix metalloproteinases (MMPs), tissue serine proteases, and cathepsins. The goal of this research is to develop an Fe/Fe3O4 nanoparticle-based system for clinical diagnostics, which has the potential to measure the activity of cancer-associated proteases in biospecimens. Nanoparticle-based "light switches" for measuring protease activity consist of fluorescent cyanine dyes and porphyrins that are attached to Fe/Fe3O4 nanoparticles via consensus sequences. These consensus sequences can be cleaved in the presence of the correct protease, thus releasing a fluorescent dye from the Fe/Fe3O4 nanoparticle, resulting in highly sensitive (down to 1 × 10(-16) mol l(-1) for 12 proteases), selective, and fast nanoplatforms (required time: 60 min).

    View details for DOI 10.1039/c3pp50260k

    View details for Web of Science ID 000333091800015

    View details for PubMedID 24096539

  • A Hybrid Soft Solar Cell Based on the Mycobacterial Porin MspA Linked to a Sensitizer-Viologen Diad JOURNAL OF THE AMERICAN CHEMICAL SOCIETY Perera, A. S., Subbaiyan, N. K., Kalita, M., Wendel, S. O., Samarakoon, T. N., D'Souza, F., Bossmann, S. H. 2013; 135 (18): 6842-6845

    Abstract

    A prototype of a nano solar cell containing the mycobacterial channel protein MspA has been successfully designed. MspA, an octameric transmembrane channel protein from Mycobacterium smegmatis, is one of the most stable proteins known to date. Eight Ruthenium(II) aminophenanthroline-viologen maleimide Diads (Ru-Diads) have been successfully bound to the MspA mutant MspAA96C via cysteine-maleimide bonds. MspA is known to form double layers in which it acts as nanoscopic surfactant. The nanostructured layer that is formed by (Ru-Diad)8MspA at the TiO2 electrode is photochemically active. The resulting "protein nano solar cell" features an incident photon conversion efficiency of 1% at 400 nm. This can be regarded as a proof-of-principle that stable proteins can be successfully integrated into the design of solar cells.

    View details for DOI 10.1021/ja403090x

    View details for Web of Science ID 000318839300026

    View details for PubMedID 23611424

  • Maleimide-Functionalized Photochromic Spirodihydroindolizines JOURNAL OF ORGANIC CHEMISTRY Shrestha, T. B., Kalita, M., Pokhrel, M., Liu, Y., Troyer, D. L., Turro, C., Bossmann, S. H., Duerr, H. 2013; 78 (5): 1903-1909

    Abstract

    Two photochromic spirodihydroindolizine/betaine systems for tethering to peptides and proteins via a maleimide function have been prepared. The absorption spectra of the betaines are in the red region of the visible spectrum and in the near-IR spectral domain, which are suitable energies of light for future in vivo applications. The half-times of cyclization have been determined for both DHI/betaine systems. The findings are consistent with a thermal barrier of varying size between the transoid and cisoid conformers of the betaines.

    View details for DOI 10.1021/jo301894s

    View details for Web of Science ID 000315707500025

    View details for PubMedID 23095100

  • Channel Blocking of MspA Revisited LANGMUIR Perera, A. S., Wang, H., Basel, M. T., Pokhrel, M., Gamage, P., Kalita, M., Wendel, S., Sears, B., Welideniya, D., Liu, Y., Turro, C., Troyer, D. L., Bossmann, S. H. 2013; 29 (1): 308-315

    Abstract

    Porin A from Mycobacterium smegmatis (MspA) is a highly stable, octameric channel protein, which acts as the main transporter of electrolytes across the cell membrane. MspA features a narrow, negatively charged constriction zone, allowing stable binding of various analytes thereby blocking the channel. Investigation of channel blocking of mycobacterial porins is of significance in developing alternate treatment methods for tuberculosis. The concept that ruthenium(II)quaterpyridinium complexes have the capability to act as efficient channel blockers for MspA and related porins, emerged after very high binding constants were measured by high-performance liquid chromatography and steady-state luminescence studies. Consequently, the interactions between the ruthenium(II) complex RuC2 molecules and MspA, leading to RuC2@MspA assemblies, have been studied utilizing time-resolved absorption/emission, atomic force microscopy, dynamic light scattering, ζ potential measurements, and isothermal titration calorimetry. The results obtained provide evidence for the formation of clusters/large aggregates of RuC2 and MspA. The results are of interest with respect to utilizing prospective channel blockers in porins. The combination of results from conceptually different techniques shed some light onto the chemical nature of MspA-channel blocker interactions thus contributing to the development of a paradigm for channel blocking.

    View details for DOI 10.1021/la3037296

    View details for Web of Science ID 000313305900037

    View details for PubMedID 23214433

  • Direct Synthesis of Aqueous Quantum Dots through 4,4 '-Bipyridine-Based Twin Ligand Strategy INORGANIC CHEMISTRY Kalita, M., Cingarapu, S., Roy, S., Park, S., Higgins, D., Jankowiak, R., Chikan, V., Klabunde, K. J., Bossmann, S. H. 2012; 51 (8): 4521-4526

    Abstract

    We report a new class of derivatized 4,4'-bipyridinium ligands for use in synthesizing highly fluorescent, extremely stable, water-soluble CdSe and CdTe quantum dots (QDs) for bioconjugation. We employed an evaporation-condensation technique, also known as solvated metal atom dispersion (SMAD), followed by a digestive ripening procedure. This method has been used to synthesize both metal nanoparticles and semiconductors in the gram scale with several stabilizing ligands in various solvents. The SMAD technique comprised evaporation condensation and stabilization of CdSe or CdTe in tetrahydrofuran. The as-prepared product was then digestively ripened in both water and dimethyl formamide, leading to narrowing of the particle size distributions. The ligands were synthesized by nucleophilic substitution (S(N)2) reactions using 4,4'-bipyridine as a nucleophile. Confocal microscopy images revealed the orange color of the nanocrystalline QDs with diameters of ~5 nm. The size has been confirmed by using transmission electron microscopy. As a part of our strategy, 85% of the 4,4'-bipyridinium salt was synthesized as propionic acid derivative and used to both stabilize the QDs in water and label basic amino acids and different biomarkers utilizing the carboxylic acid functional group. Fifteen percent of the 4,4'-bipyridinium salt was synthesized as N-propyl maleimide and used as a second ligand to label any protein containing the amino acid cysteine by means of a 1,4-Michael addition.

    View details for DOI 10.1021/ic202252m

    View details for Web of Science ID 000302833700018

    View details for PubMedID 22443511

  • Stem cell-based photodynamic therapy PHOTOCHEMICAL & PHOTOBIOLOGICAL SCIENCES Shrestha, T. B., Seo, G. M., Basel, M. T., Kalita, M., Wang, H., Villanueva, D., Pyle, M., Balivada, S., Rachakatla, R., Shinogle, H., Thapa, P. S., Moore, D., Troyer, D. L., Bossmann, S. H. 2012; 11 (7): 1251-1258

    Abstract

    We have transfected murine neural stem cells (NSCs) and rat umbilical cord matrix-derived stem cells (RUCMSCs) with a plasmid expressing gaussia luciferase (gLuc). These cells are engineered to secrete the luciferase. We have used gLuc containing supernatant from culturing the NSCs to perform in vitro photodynamic therapy of murine melanoma cells (B16F10), and RUCMSCs to perform in vivo PDT of lung melanomas in C57BL/6 mice. The treatment system was comprised of aminolevulic acid as a prodrug for the synthesis of the photosensitizer protoporphyrin IX, gaussia luciferase, and its' substrate coelenterazine. A significant reduction of the number of live melanoma cells in vitro and a borderline significant retardation of tumour growth in vivo was observed after coelenterazine-mediated PDT.

    View details for DOI 10.1039/c2pp05417e

    View details for Web of Science ID 000305533100013

    View details for PubMedID 22565929

  • MspA porin-gold nanoparticle assemblies: Enhanced binding through a controlled cysteine mutation NANO LETTERS Dani, R., Kang, M., Kalita, M., Smith, P. E., Bossmann, S. H., Chikan, V. 2008; 8 (4): 1229-1236

    Abstract

    In this study, the interactions of two gold nanoparticles of different sizes (average diameters of 3.7 +/- 2.6 and 17 +/- 3 nm) with octameric mycobacterial porin A from Mycobacterium smegmatis (MspA) and a mutant of MspA featuring a cysteine mutation in position 126 (Q126C) are investigated. From the observation of enhanced photoluminescence quenching, it is inferred that the presence of eight cysteines in the MspA Q126C mutant significantly enhances the binding of selected small gold nanoparticles within the inner pore of MspA. The large gold nanoparticle/porin complex shows photoluminescence enhancement, which is expected since the larger nanoparticles cannot dock within the homopore of MspA due to size exclusion. In addition to the fluorescence experiments, observation of energy transfer from the small gold nanoparticles to the MspA shows the close proximity of the small gold nanoparticles with the porin. Interestingly, the energy transfer of the large nanoparticle/MspA complex is completely missing. From high-performance liquid chromatography data, the estimated binding constants for small Au@MspA, large Au@MspA, small Au@MspAcys, and large Au@MspAcys are 1.3 x 10 (9), 2.22 x 10 (10), > 10 (12) (irreversible), and 1.7 x 10 (10), respectively.

    View details for DOI 10.1021/nl072658h

    View details for Web of Science ID 000254911200045

    View details for PubMedID 18318505