64 results on '"Daniel J. Shiwarski"'
Search Results
52. Graphene Microelectrode Arrays for Electrical and Optical Measurements of Human Stem Cell-Derived Cardiomyocytes
- Author
-
Sahil K. Rastogi, Guruprasad Raghavan, Jacqueline M. Bliley, Adam W. Feinberg, Daniel J. Shiwarski, and Tzahi Cohen-Karni
- Subjects
0301 basic medicine ,Bioelectronics ,Materials science ,Biocompatibility ,Graphene ,Nanotechnology ,02 engineering and technology ,021001 nanoscience & nanotechnology ,Electrochemistry ,General Biochemistry, Genetics and Molecular Biology ,Article ,law.invention ,03 medical and health sciences ,symbols.namesake ,Microelectrode ,030104 developmental biology ,Calcium imaging ,law ,Modeling and Simulation ,symbols ,0210 nano-technology ,Raman spectroscopy ,Microfabrication - Abstract
INTRODUCTION: Cell–cell communication plays a pivotal role in biological systems’ coordination and function. Electrical properties have been linked to specification and differentiation of stem cells into targeted progeny, such as neurons and cardiomyocytes. Currently, there is a critical need in developing new ways to complement fluorescent indicators, such as Ca(2+)-sensitive dyes, for direct electrophysiological measurements of cells and tissue. Here, we report a unique transparent and biocompatible graphene-based electrical platform that enables electrical and optical investigation of human embryonic stem cell-derived cardiomyocytes’ (hESC-CMs) intracellular processes and intercellular communication. METHODS: Graphene, a honeycomb sp(2) hybridized two-dimensional carbon lattice, was synthesized using low pressure chemical vapor deposition system, and was tested for biocompatibility. Au and graphene microelectrode arrays (MEAs) were fabricated using well-established microfabrication methods. Au and graphene MEAs were interfaced with hESC-CMs to perform both optical and electrical recordings. RESULTS: Optical imaging and Raman spectroscopy confirmed the presence of monolayer graphene. Viability assay showed biocompatibility of graphene. Electrochemical characterization proved graphene’s functional activity. Nitric acid treatment further enhanced the electrochemical properties of graphene. Graphene electrodes’ transparency enabled both optical and electrical recordings from hESC-CMs. Graphene MEA detected changes in beating frequency and field potential duration upon β-adrenergic receptor agonist treatment. CONCLUSION: The transparent graphene platform enables the investigation of both intracellular and intercellular communication processes and will create new avenues for bidirectional communication (sensing and stimulation) with electrically active tissues and will set the ground for investigations reported diseases such as Alzheimer, Parkinson’s disease and arrhythmias. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s12195-018-0525-z) contains supplementary material, which is available to authorized users.
- Published
- 2018
53. Potassium-regulated distal tubule WNK bodies are kidney-specific WNK1 dependent
- Author
-
Lubika J. Nkashama, Roderick J. Tan, Donna B. Stolz, Kara L. McClain, Cary R. Boyd-Shiwarski, Thomas R. Kleyman, Hima N. Namboodiri, Manojkumar A. Puthenveedu, Chou Long Huang, Allison L. Marciszyn, Jian Xie, Daniel J. Shiwarski, Ankita Roy, and Arohan R. Subramanya
- Subjects
0301 basic medicine ,Mice, 129 Strain ,030232 urology & nephrology ,Biology ,Kidney ,03 medical and health sciences ,Mice ,0302 clinical medicine ,WNK Lysine-Deficient Protein Kinase 1 ,Organelle ,medicine ,Animals ,Humans ,Distal convoluted tubule ,Microscopy, Immunoelectron ,Molecular Biology ,Mice, Knockout ,urogenital system ,HEK 293 cells ,Kidney metabolism ,Cell Biology ,Exons ,WNK1 ,Cell biology ,Mice, Inbred C57BL ,Cytosol ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Potassium ,Signal transduction ,Hydrophobic and Hydrophilic Interactions ,Homeostasis ,Signal Transduction - Abstract
With-no-lysine (WNK) kinases coordinate volume and potassium homeostasis by regulating renal tubular electrolyte transport. In the distal convoluted tubule (DCT), potassium imbalance causes WNK signaling complexes to concentrate into large discrete foci, which we call “WNK bodies.” Although these structures have been reported previously, the mechanisms that drive their assembly remain obscure. Here, we show that kidney-specific WNK1 (KS-WNK1), a truncated kinase-defective WNK1 isoform that is highly expressed in the DCT, is critical for WNK body formation. While morphologically distinct WNK bodies were evident in the distal tubules of mice subjected to dietary potassium loading and restriction, KS-WNK1 knockout mice were deficient in these structures under identical conditions. Combining in vivo observations in kidney with reconstitution studies in cell culture, we found that WNK bodies are dynamic membraneless foci that are distinct from conventional organelles, colocalize with the ribosomal protein L22, and cluster the WNK signaling pathway. The formation of WNK bodies requires an evolutionarily conserved cysteine-rich hydrophobic motif harbored within a unique N-terminal exon of KS-WNK1. We propose that WNK bodies are not pathological aggregates, but rather are KS-WNK1–dependent microdomains of the DCT cytosol that modulate WNK signaling during physiological shifts in potassium balance.
- Published
- 2017
54. TMEM16A/ANO1 Inhibits Apoptosis Via Downregulation of Bim Expression
- Author
-
Scott M. Kulich, Carolyn Kemp, Mukund Seshadri, Neal R. Godse, Roberto Gomez-Casal, Raja S. Seethala, Nayel I. Khan, Timothy F. Burns, Daniel J. Shiwarski, Umamaheswar Duvvuri, and Zachary A. Yochum
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Cell ,Down-Regulation ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Viability assay ,Anoctamin-1 ,Bcl-2-Like Protein 11 ,Cancer ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,Tumor progression ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer research ,Carcinoma, Squamous Cell ,Female ,Cisplatin - Abstract
Purpose: TMEM16A is a calcium-activated chloride channel that is amplified in a variety of cancers, including 30% of head and neck squamous cell carcinomas (HNSCCs), raising the possibility of an anti-apoptotic role in malignant cells. This study investigated this using a multimodal, translational investigation. Experimental Design: Combination of (i) in vitro HNSCC cell culture experiments assessing cell viability, apoptotic activation, and protein expression (ii) in vivo studies assessing similar outcomes, and (iii) molecular and staining analysis of human HNSCC samples. Results: TMEM16A expression was found to correlate with greater tumor size, increased Erk 1/2 activity, less Bim expression, and less apoptotic activity overall in human HNSCC. These findings were corroborated in subsequent in vitro and in vivo studies and expanded to include a cisplatin-resistant phenotype with TMEM16A overexpression. A cohort of 41 patients with laryngeal cancer demonstrated that cases that recurred after chemoradiation failure were associated with a greater TMEM16A overexpression rate than HNSCC that did not recur. Conclusions: Ultimately, this study implicates TMEM16A as a contributor to tumor progression by limiting apoptosis and as a potential biomarker of more aggressive disease. Clin Cancer Res; 23(23); 7324–32. ©2017 AACR.
- Published
- 2017
55. High dynamic range proteome imaging with the structured illumination gel imager
- Author
-
Jonathan S. Minden, Frederick Lanni, Phu T. Van, Daniel J. Shiwarski, and Victor Bass
- Subjects
Materials science ,Dynamic range ,Clinical Biochemistry ,Detector ,Analytical chemistry ,Structured illumination ,Proteomics ,Biochemistry ,Fluorescence ,Analytical Chemistry ,Electrophoresis ,Proteome ,Biological system ,High dynamic range - Abstract
A current challenge for proteomics is detecting proteins over the large concentration ranges found in complex biological samples such as whole-cell extracts. Currently, no unbiased, whole-proteome analysis scheme is capable of detecting the full range of cellular proteins. This is due in part to the limited dynamic range of the detectors used to sense proteins or peptides. We present a new technology, structured illumination (SI) gel imager, which detects fluorescently labeled proteins in electrophoretic gels over a 1 000 000-fold concentration range. SI uses computer-generated masks to attenuate the illumination of highly abundant proteins, allowing for long exposures of low-abundance proteins, thus avoiding detector saturation. A series of progressively masked gel images are assembled into a single, very high dynamic range image. We demonstrate that the SI imager can detect proteins over a concentration range of approximately 1 000 000-fold, making it a useful tool for comprehensive, unbiased proteome-wide surveys.
- Published
- 2014
- Full Text
- View/download PDF
56. PI3K class II α regulates δ-opioid receptor export from the
- Author
-
Daniel J, Shiwarski, Marlena, Darr, Cheryl A, Telmer, Marcel P, Bruchez, and Manojkumar A, Puthenveedu
- Subjects
Neurons ,Cell Membrane ,Articles ,Phosphatidylinositols ,PC12 Cells ,Rats ,Phosphatidylinositol 3-Kinases ,Protein Transport ,Membrane Trafficking ,Receptors, Opioid, delta ,Nerve Growth Factor ,Animals ,Phosphorylation ,Cells, Cultured ,Signal Transduction ,trans-Golgi Network - Abstract
The δ-opioid receptor (δR) is retained in intracellular structures in neurons, but the mechanisms of retention and regulated export are not known. The atypical phosphoinositide-3 kinase C2A is required and sufficient for NGF-regulated δR export from the trans-Golgi network and surface transport., The interplay between signaling and trafficking by G protein–coupled receptors (GPCRs) has focused mainly on endocytic trafficking. Whether and how surface delivery of newly synthesized GPCRs is regulated by extracellular signals is less understood. Here we define a signaling-regulated checkpoint at the trans-Golgi network (TGN) that controls the surface delivery of the delta opioid receptor (δR). In PC12 cells, inhibition of phosphoinositide-3 kinase (PI3K) activity blocked export of newly synthesized δR from the Golgi and delivery to the cell surface, similar to treatment with nerve growth factor (NGF). Depletion of class II phosphoinositide-3 kinase α (PI3K C2A), but not inhibition of class I PI3K, blocked δR export to comparable levels and attenuated δR-mediated cAMP inhibition. NGF treatment displaced PI3K C2A from the Golgi and optogenetic recruitment of the PI3K C2A kinase domain to the TGN-induced δR export downstream of NGF. Of importance, PI3K C2A expression promotes export of endogenous δR in primary trigeminal ganglion neurons. Taken together, our results identify PI3K C2A as being required and sufficient for δR export and surface delivery in neuronal cells and suggest that it could be a key modulator of a novel Golgi export checkpoint that coordinates GPCR delivery to the surface.
- Published
- 2017
57. DOG1: a novel marker of salivary acinar and intercalated duct differentiation
- Author
-
Jacinthe Chenevert, Daniel J. Shiwarski, Umamaheswar Duvvuri, Kathleen Cieply, Simion I. Chiosea, Raja R. Seethala, Jean Kim, and Sanja Dacic
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Acinar Cells ,Biology ,Pathology and Forensic Medicine ,Acinic cell carcinoma ,Chloride Channels ,Biomarkers, Tumor ,medicine ,Humans ,Salivary Ducts ,RNA, Messenger ,Anoctamin-1 ,In Situ Hybridization, Fluorescence ,Salivary gland ,Carcinoma, Acinar Cell ,Reverse Transcriptase Polymerase Chain Reaction ,Carcinoma ,Myoepithelial cell ,Cell Differentiation ,Intercalated duct ,Salivary Gland Neoplasms ,medicine.disease ,Immunohistochemistry ,Neoplasm Proteins ,Staining ,Serous fluid ,Cell Transformation, Neoplastic ,medicine.anatomical_structure - Abstract
Anoctamin-1 (ANO1) (DOG1, TMEM16a) is a calcium-activated chloride channel initially described in gastrointestinal stromal tumors, but now known to be expressed in a variety of normal and tumor tissues including salivary tissue in murine models. We herein perform a comprehensive survey of DOG1 expression in 156 cases containing non-neoplastic human salivary tissues and tumors. ANO1 mRNA levels were significantly higher (8-fold increase, P
- Published
- 2012
- Full Text
- View/download PDF
58. TMEM16A Induces MAPK and Contributes Directly to Tumorigenesis and Cancer Progression
- Author
-
Xin Huang, Brian D. Harfe, Rainer Schreiber, Jennifer R. Grandis, Susanne M. Gollin, Daniel J. Shiwarski, Ann Marie Egloff, Dong Xiao, Umamaheswar Duvvuri, Raja S. Seethala, Jason R. Rock, Carol A. Bertrand, Xing Chen, Robert S. Edinger, Brian J. Henson, Vivian Wai Yan Lui, and Karl Kunzelmann
- Subjects
MAPK/ERK pathway ,Cancer Research ,Biology ,medicine.disease_cause ,Article ,Cell Line ,Mice ,Cyclin D1 ,Chloride Channels ,medicine ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,Anoctamin-1 ,In Situ Hybridization, Fluorescence ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Kinase ,Cancer ,medicine.disease ,Head and neck squamous-cell carcinoma ,Neoplasm Proteins ,Cell biology ,Enzyme Activation ,Cell Transformation, Neoplastic ,Oncology ,Head and Neck Neoplasms ,Cell culture ,Enzyme Induction ,Carcinoma, Squamous Cell ,Disease Progression ,Cancer research ,Mitogen-Activated Protein Kinases ,Carcinogenesis ,Cell Division - Abstract
Frequent gene amplification of the receptor-activated calcium-dependent chloride channel TMEM16A (TAOS2 or ANO1) has been reported in several malignancies. However, its involvement in human tumorigenesis has not been previously studied. Here, we show a functional role for TMEM16A in tumor growth. We found TMEM16A overexpression in 80% of head and neck squamous cell carcinoma (SCCHN), which correlated with decreased overall survival in patients with SCCHN. TMEM16A overexpression significantly promoted anchorage-independent growth in vitro, and loss of TMEM16A resulted in inhibition of tumor growth both in vitro and in vivo. Mechanistically, TMEM16A-induced cancer cell proliferation and tumor growth were accompanied by an increase in extracellular signal–regulated kinase (ERK)1/2 activation and cyclin D1 induction. Pharmacologic inhibition of MEK/ERK and genetic inactivation of ERK1/2 (using siRNA and dominant-negative constructs) abrogated the growth effect of TMEM16A, indicating a role for mitogen-activated protein kinase (MAPK) activation in TMEM16A-mediated proliferation. In addition, a developmental small-molecule inhibitor of TMEM16A, T16A-inh01 (A01), abrogated tumor cell proliferation in vitro. Together, our findings provide a mechanistic analysis of the tumorigenic properties of TMEM16A, which represents a potentially novel therapeutic target. The development of small-molecule inhibitors against TMEM16A may be clinically relevant for treatment of human cancers, including SCCHN. Cancer Res; 72(13); 3270–81. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
59. Distinct G protein-coupled receptor recycling pathways allow spatial control of downstream G protein signaling
- Author
-
Shanna L. Bowman, Manojkumar A. Puthenveedu, and Daniel J. Shiwarski
- Subjects
0301 basic medicine ,Gs alpha subunit ,Retromer ,Endosome ,G protein ,Endosomes ,Biology ,03 medical and health sciences ,Phosphoserine ,Membrane Microdomains ,Protein Domains ,GTP-Binding Proteins ,Report ,Cyclic AMP ,Humans ,Protein kinase A ,Research Articles ,G protein-coupled receptor ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Endocytosis ,Cell biology ,Sorting nexin ,030104 developmental biology ,HEK293 Cells ,Receptors, Adrenergic, beta-2 ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
GPCRs can activate different programs of gene expression from the plasma membrane and the endosome. Bowman et al. show that signaling by endosomal β-2 adrenergic receptors occurs at the microdomains that GPCRs use for sequence-dependent recycling., G protein–coupled receptors (GPCRs) are recycled via a sequence-dependent pathway that is spatially and biochemically distinct from bulk recycling. Why there are two distinct recycling pathways from the endosome is a fundamental question in cell biology. In this study, we show that the separation of these two pathways is essential for normal spatial encoding of GPCR signaling. The prototypical β-2 adrenergic receptor (B2AR) activates Gα stimulatory protein (Gαs) on the endosome exclusively in sequence-dependent recycling tubules marked by actin/sorting nexin/retromer tubular (ASRT) microdomains. B2AR was detected in an active conformation in bulk recycling tubules, but was unable to activate Gαs. Protein kinase A phosphorylation of B2AR increases the fraction of receptors localized to ASRT domains and biases the downstream transcriptional effects of B2AR to genes controlled by endosomal signals. Our results identify the physiological relevance of separating GPCR recycling from bulk recycling and suggest a mechanism to tune downstream responses of GPCR signaling by manipulating the spatial origin of G protein signaling.
- Published
- 2015
60. To 'Grow' or 'Go': TMEM16A Expression as a Switch between Tumor Growth and Metastasis in SCCHN
- Author
-
Carol A. Bertrand, Jeffrey N. Myers, Chunbo Shao, Dong Xiao, Raja R. Seethala, L. Alex Gaither, Daniel J. Shiwarski, Jean Kim, Manojkumar A. Puthenveedu, Umamaheswar Duvvuri, Patrick K. Ha, Daisuke Sano, Anke Bill, and Jennifer R. Grandis
- Subjects
Cancer Research ,Pathology ,Carcinogenesis ,medicine.disease_cause ,Metastasis ,Mice ,Cell Movement ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Regulation of gene expression ,Tumor ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,Head and Neck Neoplasms ,Lymphatic Metastasis ,DNA methylation ,Carcinoma, Squamous Cell ,69999 Biological Sciences not elsewhere classified ,Biotechnology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Oncology and Carcinogenesis ,Biology ,Article ,Cell Line ,Rare Diseases ,Chloride Channels ,Cell Line, Tumor ,Genetics ,Carcinoma ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Epithelial–mesenchymal transition ,Dental/Oral and Craniofacial Disease ,Anoctamin-1 ,Cell Proliferation ,Neoplastic ,Cell growth ,Squamous Cell Carcinoma of Head and Neck ,Membrane Proteins ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,Cytoskeletal Proteins ,Squamous Cell ,Gene Expression Regulation ,FOS: Biological sciences ,Cancer research - Abstract
Purpose: Tumor metastasis is the leading cause of death in patients with cancer. However, the mechanisms that underlie metastatic progression remain unclear. We examined TMEM16A (ANO1) expression as a key factor shifting tumors between growth and metastasis. Experimental Design: We evaluated 26 pairs of primary and metastatic lymph node (LN) tissue from patients with squamous cell carcinoma of the head and neck (SCCHN) for differential expression of TMEM16A. In addition, we identified mechanisms by which TMEM16A expression influences tumor cell motility via proteomic screens of cell lines and in vivo mouse studies of metastasis. Results: Compared with primary tumors, TMEM16A expression decreases in metastatic LNs of patients with SCCHN. Stable reduction of TMEM16A expression enhances cell motility and increases metastases while decreasing tumor proliferation in an orthotopic mouse model. Evaluation of human tumor tissues suggests an epigenetic mechanism for decreasing TMEM16A expression through promoter methylation that correlated with a transition between an epithelial and a mesenchymal phenotype. These effects of TMEM16A expression on tumor cell size and epithelial-to-mesenchymal transition (EMT) required the amino acid residue serine 970 (S970); however, mutation of S970 to alanine does not disrupt the proliferative advantages of TMEM16A overexpression. Furthermore, S970 mediates the association of TMEM16A with Radixin, an actin-scaffolding protein implicated in EMT. Conclusions: Together, our results identify TMEM16A, an eight transmembrane domain Ca2+-activated Cl− channel, as a primary driver of the “Grow” or “Go” model for cancer progression, in which TMEM16A expression acts to balance tumor proliferation and metastasis via its promoter methylation. Clin Cancer Res; 20(17); 4673–88. ©2014 AACR.
- Published
- 2014
61. Cell-autonomous regulation of Mu-opioid receptor recycling by substance P
- Author
-
Daniel J. Shiwarski, Stefan Schulz, Shanna L. Bowman, Manojkumar A. Puthenveedu, Amynah A. Pradhan, and Amanda L. Soohoo
- Subjects
Male ,Nociception ,medicine.medical_specialty ,medicine.drug_class ,Receptors, Opioid, mu ,Endogeny ,Substance P ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Trigeminal ganglion ,Mice ,0302 clinical medicine ,Opioid receptor ,Internal medicine ,medicine ,Animals ,Humans ,Neurotransmitter ,Receptor ,lcsh:QH301-705.5 ,Protein kinase C ,Cells, Cultured ,Protein Kinase C ,030304 developmental biology ,Neurons ,0303 health sciences ,Morphine ,Receptors, Neurokinin-1 ,Endocytosis ,Cell biology ,Rats ,Analgesics, Opioid ,Fentanyl ,Mice, Inbred C57BL ,Protein Transport ,Endocrinology ,HEK293 Cells ,chemistry ,lcsh:Biology (General) ,nervous system ,Trigeminal Ganglion ,μ-opioid receptor ,030217 neurology & neurosurgery - Abstract
Summary How neurons coordinate and reprogram multiple neurotransmitter signals is an area of broad interest. Here, we show that substance P (SP), a neuropeptide associated with inflammatory pain, reprograms opioid receptor recycling and signaling. SP, through activation of the neurokinin 1 (NK1R) receptor, increases the post-endocytic recycling of the mu-opioid receptor (MOR) in trigeminal ganglion (TG) neurons in an agonist-selective manner. SP-mediated protein kinase C (PKC) activation is both required and sufficient for increasing recycling of exogenous and endogenous MOR in TG neurons. The target of this cross-regulation is MOR itself, given that mutation of either of two PKC phosphorylation sites on MOR abolishes the SP-induced increase in recycling and resensitization. Furthermore, SP enhances the resensitization of fentanyl-induced, but not morphine-induced, antinociception in mice. Our results define a physiological pathway that cross-regulates opioid receptor recycling via direct modification of MOR and suggest a mode of homeostatic interaction between the pain and analgesic systems.
- Published
- 2014
62. Effects of TMEM16A Expression on Motility and Metastasis in Epithelial Tumor Cells
- Author
-
Suzanne Gollin, Jennifer R. Grandis, Xin Huang, Lu Mei He, Daniel J. Shiwarski, and Umamaheswar Duvvuri
- Subjects
Genetics ,Cancer research ,medicine ,Motility ,Tumor cells ,Biology ,medicine.disease ,Molecular Biology ,Biochemistry ,Biotechnology ,Metastasis - Published
- 2010
- Full Text
- View/download PDF
63. Abstract LB-220: TMEM16A, a novel calcium-activated chloride channel, modulates tumor proliferation via MAPK and Cyclin-D1 signaling
- Author
-
Ann Marie Egloff, Carol A. Bertrand, Xin Huang, Jennifer R. Grandis, Raja R. Seethala, Susanne M. Gollin, Daniel J. Shiwarski, and Umamaheswar Duvvuri
- Subjects
MAPK/ERK pathway ,Cancer Research ,Gene knockdown ,Stromal cell ,business.industry ,Cancer ,medicine.disease ,Cyclin D1 ,Oncology ,Cancer cell ,Immunology ,Cancer research ,Medicine ,Signal transduction ,business ,Chloride channel activity - Abstract
TMEM16A, a calcium-activated chloride channel, was recently isolated from oral cavity cancer cells. TMEM16A is amplified in head and neck cancers and overexpressed in gastrointestinal stromal tumors (GIST's); however, the role that TMEM16A plays in promoting tumor growth remains unclear. The goal of this research was to establish the biologic function of TMEM16A in the context of epithelial malignancies. We hypothesized that TMEM16A contributes to tumor cell survival and may be a useful prognostic factor for patients with squamous cell carcinoma of the head and neck (SCCHN). Knockdown and overexpressing SCCHN and T24 cell lines were transduced with lentiviral particles encoding RNAi and engineered to stably overexpress TMEM16A. For functional verification, electrophysiologic assays were performed to establish that the altered TMEM16A expression was modulating the chloride current. To elucidate a mechanism of TMEM16A's proliferative effects, immunoblotting and pharmacologic inhibition of potential downstream targets were performed. Our results demonstrate that the levels of TMEM16A expression modulates proliferation in vitro and in vivo, and that stable overexpression of TMEM16A increases resistance to cisplatin therapy as compared to cancer cell lines expressing endogenous TMEM16A. Specificity of these phenotypic results were confirmed using genetic rescue experiments. Next, we identified that the MAPK/Cyclin-D1 axis was a critical signaling pathway in TMEM16A-induced proliferation, and that pMAPK/MAPK and Cyclin-D1 levels paralleled expression of TMEM16A. Finally, using pharmacologic agents niflumic acid or NPPB to inhibit the TMEM16A chloride channel activity in combination with a MAPK inhibitor (U0126) demonstrated additive effects significantly decreasing cancer cell proliferation in vitro. In summary, our results provide novel insights into the biologic function of TMEM16A and its role in tumor cell proliferation. This research has enhanced our understanding about the pathogenesis of SCCHN tumors, and can potentially be translated to many other cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-220. doi:10.1158/1538-7445.AM2011-LB-220
- Published
- 2011
- Full Text
- View/download PDF
64. Dynamic loading of human engineered heart tissue enhances contractile function and drives a desmosome-linked disease phenotype
- Author
-
Daniel J. Shiwarski, Andrew H. Lee, Martijn F Hoes, Rebecca M. Duffy, Peter van der Meer, Yan Sun, Rachelle N. Palchesko, Duco Kramer, Maria C. Bolling, Anna Kalmykov, Alexander S Teplenin, Rudolf A. de Boer, Albert J. H. Suurmeijer, Brian Coffin, Nils Bomer, Mathilde C.S.C. Vermeer, Joshua W. Tashman, Adam W. Feinberg, Ivan Batalov, Jan D. H. Jongbloed, Daniël A. Pijnappels, L Volkers, Jacqueline M. Bliley, Cardiovascular Centre (CVC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Restoring Organ Function by Means of Regenerative Medicine (REGENERATE)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cardiac output ,Induced Pluripotent Stem Cells ,Cardiomyopathy ,Diastole ,030204 cardiovascular system & hematology ,Contractility ,03 medical and health sciences ,0302 clinical medicine ,Afterload ,Internal medicine ,medicine ,Myocyte ,Humans ,Myocytes, Cardiac ,Heart formation ,Tissue Engineering ,Chemistry ,General Medicine ,Desmosomes ,medicine.disease ,Myocardial Contraction ,Preload ,030104 developmental biology ,Phenotype ,Cardiology - Abstract
The role that mechanical forces play in shaping the structure and function of the heart is critical to understanding heart formation and the etiology of disease but is challenging to study in patients. Engineered heart tissues (EHTs) incorporating human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have the potential to provide insight into these adaptive and maladaptive changes. However, most EHT systems cannot model both preload (stretch during chamber filling) and afterload (pressure the heart must work against to eject blood). Here, we have developed a new dynamic EHT (dyn-EHT) model that enables us to tune preload and have unconstrained contractile shortening of >10%. To do this, three-dimensional (3D) EHTs were integrated with an elastic polydimethylsiloxane strip providing mechanical preload and afterload in addition to enabling contractile force measurements based on strip bending. Our results demonstrated that dynamic loading improves the function of wild-type EHTs on the basis of the magnitude of the applied force, leading to improved alignment, conduction velocity, and contractility. For disease modeling, we used hiPSC-derived cardiomyocytes from a patient with arrhythmogenic cardiomyopathy due to mutations in the desmoplakin gene. We demonstrated that manifestation of this desmosome-linked disease state required dyn-EHT conditioning and that it could not be induced using 2D or standard 3D EHT approaches. Thus, a dynamic loading strategy is necessary to provoke the disease phenotype of diastolic lengthening, reduction of desmosome counts, and reduced contractility, which are related to primary end points of clinical disease, such as chamber thinning and reduced cardiac output.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.