Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Value Health ; 25(10): 1760-1767, 2022 10.
Article in English | MEDLINE | ID: mdl-35595634

ABSTRACT

OBJECTIVES: The Innovative Medicines Initiative-funded, multistakeholders project Healthcare Alliance for Resourceful Medicine Offensive Against Neoplasms in Hematology (HARMONY) created a task force involving patient organizations, medical associations, pharmaceutical companies, and health technology assessment/regulator agencies' representatives to evaluate the suitability of previously established value frameworks (VFs) for assessing the clinical and societal impact of new interventions for hematologic malignancies (HMs). METHODS: Since the HARMONY stakeholders identified the inclusion of patients' points of view on evaluating VFs as a priority, surveys were conducted with the patient organizations active in HMs and part of the HARMONY network, together with key opinion leaders, pharmaceutical companies, and regulators, to establish which outcomes were important for each HM. Next, to evaluate VFs against the sources of information taken into account (randomized clinical trials, registries, real-world data), structured questionnaires were created and filled by HARMONY health professionals to specify preferred data sources per malignancy. Finally, a framework evaluation module was built to analyze existing clinical VFs (American Society of Clinical Oncology, European Society of Medical Oncology, Magnitude of Clinical Benefit Scale, Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen, Institute for Clinical and Economic Review, National Comprehensive Cancer Network Evidence Blocks, and patient-perspective VF). RESULTS: The comparative analysis describes challenges and opportunities for the use of each framework in the context of HMs and drafts possible lines of action for creating or integrating a more specific, patient-focused clinical VF for HMs. CONCLUSIONS: None of the frameworks meets the HARMONY goals for a tool that applies to HMs and assesses in a transparent, reproducible, and systematic way the therapeutic value of innovative health technologies versus available alternatives, taking a patient-centered approach and using real-world evidence.


Subject(s)
Hematologic Neoplasms , Hematology , Neoplasms , Health Resources , Hematologic Neoplasms/therapy , Humans , Neoplasms/therapy , Pharmaceutical Preparations
2.
Pain ; 160(10): 2305-2315, 2019 10.
Article in English | MEDLINE | ID: mdl-31365468

ABSTRACT

Nerve growth factor (NGF) and its receptors TrkA and p75 play a key role in the development and function of peripheral nociceptive neurons. Here, we describe novel technology to selectively photoablate TrkA-positive nociceptors through delivery of a phototoxic agent coupled to an engineered NGF ligand and subsequent near-infrared illumination. We demonstrate that this approach allows for on demand and localized reversal of pain behaviors in mouse models of acute, inflammatory, neuropathic, and joint pain. To target peripheral nociceptors, we generated a SNAP-tagged NGF derivative NGF that binds to TrkA/p75 receptors but does not provoke signaling in TrkA-positive cells or elicit pain behaviors in mice. NGF was coupled to the photosensitizer IRDye700DX phthalocyanine (IR700) and injected subcutaneously. After near-infrared illumination of the injected area, behavioral responses to nociceptive mechanical and sustained thermal stimuli, but not innocuous stimuli, were substantially reduced. Similarly, in models of inflammatory, osteoarthritic, and neuropathic pain, mechanical hypersensitivity was abolished for 3 weeks after a single treatment regime. We demonstrate that this loss of pain behavior coincides with the retraction of neurons from the skin which then reinnervate the epidermis after 3 weeks corresponding with the return of mechanical hypersensitivity. Thus NGF-mediated photoablation is a minimally invasive approach to reversibly silence nociceptor input from the periphery, and control pain and hypersensitivity to mechanical stimuli.


Subject(s)
Ablation Techniques/methods , Nerve Growth Factor/administration & dosage , Neuralgia/therapy , Nociceptors/drug effects , Pain Measurement/methods , Photosensitizing Agents/administration & dosage , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuralgia/physiopathology , Nociceptors/physiology , PC12 Cells , Rats
3.
Nat Commun ; 9(1): 1640, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691410

ABSTRACT

Mechanical allodynia is a major symptom of neuropathic pain whereby innocuous touch evokes severe pain. Here we identify a population of peripheral sensory neurons expressing TrkB that are both necessary and sufficient for producing pain from light touch after nerve injury in mice. Mice in which TrkB-Cre-expressing neurons are ablated are less sensitive to the lightest touch under basal conditions, and fail to develop mechanical allodynia in a model of neuropathic pain. Moreover, selective optogenetic activation of these neurons after nerve injury evokes marked nociceptive behavior. Using a phototherapeutic approach based upon BDNF, the ligand for TrkB, we perform molecule-guided laser ablation of these neurons and achieve long-term retraction of TrkB-positive neurons from the skin and pronounced reversal of mechanical allodynia across multiple types of neuropathic pain. Thus we identify the peripheral neurons which transmit pain from light touch and uncover a novel pharmacological strategy for its treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyperalgesia/therapy , Laser Therapy , Membrane Glycoproteins/metabolism , Neuralgia/metabolism , Neuralgia/therapy , Protein-Tyrosine Kinases/metabolism , Sensory Receptor Cells/radiation effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Ligands , Male , Membrane Glycoproteins/genetics , Mice , Neuralgia/genetics , Neuralgia/physiopathology , Protein-Tyrosine Kinases/genetics , Sensory Receptor Cells/metabolism , Touch/radiation effects
4.
Elife ; 52016 12 13.
Article in English | MEDLINE | ID: mdl-27976998

ABSTRACT

At its most fundamental level, touch sensation requires the translation of mechanical energy into mechanosensitive ion channel opening, thereby generating electro-chemical signals. Our understanding of this process, especially how the cytoskeleton influences it, remains unknown. Here we demonstrate that mice lacking the α-tubulin acetyltransferase Atat1 in sensory neurons display profound deficits in their ability to detect mechanical stimuli. We show that all cutaneous afferent subtypes, including nociceptors have strongly reduced mechanosensitivity upon Atat1 deletion, and that consequently, mice are largely insensitive to mechanical touch and pain. We establish that this broad loss of mechanosensitivity is dependent upon the acetyltransferase activity of Atat1, which when absent leads to a decrease in cellular elasticity. By mimicking α-tubulin acetylation genetically, we show both cellular rigidity and mechanosensitivity can be restored in Atat1 deficient sensory neurons. Hence, our results indicate that by influencing cellular stiffness, α-tubulin acetylation sets the force required for touch.


Subject(s)
Acetyltransferases/metabolism , Neurons, Afferent/enzymology , Neurons, Afferent/physiology , Protein Processing, Post-Translational , Touch , Tubulin/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Gene Deletion , Mice , Microtubule Proteins
5.
EMBO Rep ; 17(4): 585-600, 2016 04.
Article in English | MEDLINE | ID: mdl-26929027

ABSTRACT

Itch, the unpleasant sensation that elicits a desire to scratch, is mediated by specific subtypes of cutaneous sensory neuron. Here, we identify a subpopulation of itch-sensing neurons based on their expression of the receptor tyrosine kinase Ret. We apply flow cytometry to isolate Ret-positive neurons from dorsal root ganglia and detected a distinct population marked by low levels of Ret and absence of isolectin B4 binding. We determine the transcriptional profile of these neurons and demonstrate that they express neuropeptides such as somatostatin (Sst), the NGF receptor TrkA, and multiple transcripts associated with itch. We validate the selective expression of Sst using an Sst-Cre driver line and ablated these neurons by generating mice in which the diphtheria toxin receptor is conditionally expressed from the sensory neuron-specific Avil locus. Sst-Cre::Avil(iDTR) mice display normal nociceptive responses to thermal and mechanical stimuli. However, scratching behavior evoked by interleukin-31 (IL-31) or agonist at the 5HT1F receptor is significantly reduced. Our data provide a molecular signature for a subpopulation of neurons activated by multiple pruritogens.


Subject(s)
Ganglia, Spinal/metabolism , Proto-Oncogene Proteins c-ret/genetics , Pruritus/genetics , Sensory Receptor Cells/metabolism , Somatostatin/genetics , Animals , Gene Expression Profiling , Heparin-binding EGF-like Growth Factor/genetics , In Situ Hybridization , Lectins/metabolism , Mice , Microfilament Proteins/genetics , Neurons, Afferent/metabolism , Neuropeptides/metabolism , Receptor, Nerve Growth Factor/genetics
6.
Endocr Rev ; 35(1): 106-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24311738

ABSTRACT

The Na(+)/I(-) symporter (NIS) is the plasma membrane glycoprotein that mediates active I(-) transport in the thyroid and other tissues, such as salivary glands, stomach, lactating breast, and small intestine. In the thyroid, NIS-mediated I(-) uptake plays a key role as the first step in the biosynthesis of the thyroid hormones, of which iodine is an essential constituent. These hormones are crucial for the development of the central nervous system and the lungs in the fetus and the newborn and for intermediary metabolism at all ages. Since the cloning of NIS in 1996, NIS research has become a major field of inquiry, with considerable impact on many basic and translational areas. In this article, we review the most recent findings on NIS, I(-) homeostasis, and related topics and place them in historical context. Among many other issues, we discuss the current outlook on iodide deficiency disorders, the present stage of understanding of the structure/function properties of NIS, information gleaned from the characterization of I(-) transport deficiency-causing NIS mutations, insights derived from the newly reported crystal structures of prokaryotic transporters and 3-dimensional homology modeling, and the novel discovery that NIS transports different substrates with different stoichiometries. A review of NIS regulatory mechanisms is provided, including a newly discovered one involving a K(+) channel that is required for NIS function in the thyroid. We also cover current and potential clinical applications of NIS, such as its central role in the treatment of thyroid cancer, its promising use as a reporter gene in imaging and diagnostic procedures, and the latest studies on NIS gene transfer aimed at extending radioiodide treatment to extrathyroidal cancers, including those involving specially engineered NIS molecules.


Subject(s)
Iodine/deficiency , Symporters/metabolism , Thyroid Gland/metabolism , Thyroid Hormones/biosynthesis , Thyroid Neoplasms/etiology , Humans , Thyroid Neoplasms/metabolism
7.
Cancer Res ; 72(21): 5505-15, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22962269

ABSTRACT

A number of solute carrier (SLC) proteins are subject to changes in expression and activity during carcinogenesis. Whether these changes play a role in carcinogenesis is unclear, except for some nutrients and ion carriers whose deregulation ensures the necessary reprogramming of energy metabolism in cancer cells. In this study, we investigated the functional role in tumor progression of the sodium/iodide symporter (NIS; aka SLC5A5), which is upregulated and mislocalized in many human carcinomas. Notably, we found that NIS enhanced cell migration and invasion without ion transport being involved. These functions were mediated by NIS binding to leukemia-associated RhoA guanine exchange factor, a Rho guanine exchange factor that activates the small GTPase RhoA. Sequestering NIS in intracellular organelles or impairing its targeting to the cell surface (as observed in many cancers) led to a further increase in cell motility and invasiveness. In sum, our results established NIS as a carrier protein that interacts with a major cell signaling hub to facilitate tumor cell locomotion and invasion.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Neoplasm Invasiveness/pathology , Signal Transduction/physiology , Symporters/metabolism , Cell Line, Tumor , Cell Movement/physiology , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rho Guanine Nucleotide Exchange Factors , Transduction, Genetic , Two-Hybrid System Techniques
8.
Am J Physiol Cell Physiol ; 296(4): C654-62, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19052257

ABSTRACT

Absorption of dietary iodide, presumably in the small intestine, is the first step in iodide (I(-)) utilization. From the bloodstream, I(-) is actively taken up via the Na(+)/I(-) symporter (NIS) in the thyroid for thyroid hormone biosynthesis and in such other tissues as lactating breast, which supplies I(-) to the newborn in the milk. The molecular basis for intestinal I(-) absorption is unknown. We sought to determine whether I(-) is actively accumulated by enterocytes and, if so, whether this process is mediated by NIS and regulated by I(-) itself. NIS expression was localized exclusively at the apical surface of rat and mouse enterocytes. In vivo intestine-to-blood transport of pertechnetate, a NIS substrate, was sensitive to the NIS inhibitor perchlorate. Brush border membrane vesicles accumulated I(-) in a sodium-dependent, perchlorate-sensitive manner with kinetic parameters similar to those of thyroid cells. NIS was expressed in intestinal epithelial cell line 6, and I(-) uptake in these cells was also kinetically similar to that in thyrocytes. I(-) downregulated NIS protein expression and its own NIS-mediated transport both in vitro and in vivo. We conclude that NIS is functionally expressed on the apical surface of enterocytes, where it mediates active I(-) accumulation. Therefore, NIS is a significant and possibly central component of the I(-) absorption system in the small intestine, a system of key importance for thyroid hormone biosynthesis and thus systemic intermediary metabolism.


Subject(s)
Enterocytes/metabolism , Intestinal Absorption , Intestine, Small/metabolism , Iodides/metabolism , Symporters/metabolism , Animals , Cell Line , Cell Polarity , Enterocytes/drug effects , Intestinal Absorption/drug effects , Intestine, Small/cytology , Intestine, Small/drug effects , Iodine Radioisotopes , Kinetics , Male , Mice , Microvilli/metabolism , Perchlorates/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Pertechnetate Tc 99m/metabolism , Symporters/antagonists & inhibitors , Thyroid Gland/metabolism
9.
Proc Natl Acad Sci U S A ; 104(51): 20250-5, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18077370

ABSTRACT

The Na(+)/I(-) symporter (NIS) is a key plasma membrane protein that mediates active I(-) uptake in the thyroid, lactating breast, and other tissues with an electrogenic stoichiometry of 2 Na(+) per I(-). In the thyroid, NIS-mediated I(-) uptake is the first step in the biosynthesis of the iodine-containing thyroid hormones, which are essential early in life for proper CNS development. In the lactating breast, NIS mediates the translocation of I(-) to the milk, thus supplying this essential anion to the nursing newborn. Perchlorate (ClO(4)(-)) is a well known competitive inhibitor of NIS. Exposure to food and water contaminated with ClO(4)(-) is common in the U.S. population, and the public health impact of such exposure is currently being debated. To date, it is still uncertain whether ClO(4)(-) is a NIS blocker or a transported substrate of NIS. Here we show in vitro and in vivo that NIS actively transports ClO(4)(-), including ClO(4)(-) translocation to the milk. A simple mathematical fluxes model accurately predicts the effect of ClO(4)(-) transport on the rate and extent of I(-) accumulation. Strikingly, the Na(+)/ ClO(4)(-) transport stoichiometry is electroneutral, uncovering that NIS translocates different substrates with different stoichiometries. That NIS actively concentrates ClO(4)(-) in maternal milk suggests that exposure of newborns to high levels of ClO(4)(-) may pose a greater health risk than previously acknowledged because ClO(4)(-) would thus directly inhibit the newborns' thyroidal I(-) uptake.


Subject(s)
Environmental Pollutants/metabolism , Iodine/metabolism , Perchlorates/metabolism , Symporters/metabolism , Animals , Biological Transport , Cell Line , Dogs , Iodine/analysis , Kinetics , Milk/chemistry , Models, Theoretical , Perchlorates/analysis , Rats , Transfection
10.
Am J Physiol Cell Physiol ; 292(2): C814-23, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16987991

ABSTRACT

The sodium iodide symporter (NIS) mediates iodide (I(-)) transport in the thyroid gland and other tissues and is of increasing importance as a therapeutic target and nuclear imaging reporter. NIS activity in vitro is currently measured with radiotracers and electrophysiological techniques. We report on the development of a novel live cell imaging assay of NIS activity using the I(-)-sensitive and genetically encodable yellow fluorescent protein (YFP) variant YFP-H148Q/I152L. In FRTL-5 thyrocytes stably expressing YFP-H148Q/I152L, I(-) induced a rapid and reversible decrease in cellular fluorescence characterized by 1) high affinity for extracellular I(-) (35 muM), 2) inhibition by the NIS inhibitor perchlorate, 3) extracellular Na(+) dependence, and 4) TSH dependence, suggesting that fluorescence changes are due to I(-) influx via NIS. Individual cells within a population of FRTL-5 cells exhibited a 3.5-fold variation in the rate of NIS-mediated I(-) influx, illustrating the utility of YFP-H148Q/I152L to detect cell-to-cell difference in NIS activity. I(-) also caused a perchlorate-sensitive decrease in YFP-H148Q/I152L fluorescence in COS-7 cells expressing NIS but not in cells lacking NIS. These results demonstrate that YFP-H148Q/I152L is a sensitive biosensor of NIS-mediated I(-) uptake in thyroid cells and in nonthyroidal cells following gene transfer and suggest that fluorescence detection of cellular I(-) may be a useful tool by which to study the pathophysiology and pharmacology of NIS.


Subject(s)
Biosensing Techniques , Luminescent Proteins/metabolism , Sodium/metabolism , Symporters/metabolism , Animals , Cell Line , Chlorocebus aethiops , Iodides/metabolism , Ion Transport , Luminescent Proteins/genetics , Microscopy, Fluorescence , Perchlorates/pharmacology , Rats , Rats, Inbred F344 , Symporters/antagonists & inhibitors , Thyroid Gland/cytology , Thyrotropin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...