Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Am J Transplant ; 22(2): 552-564, 2022 02.
Article in English | MEDLINE | ID: mdl-34379885

ABSTRACT

Ex vivo lung perfusion (EVLP) is a novel lung preservation strategy that facilitates the use of marginal allografts; however, it is more expensive than static cold storage (SCS). To understand how preservation method might affect postoperative costs, we compared outcomes and index hospitalization costs among matched EVLP and SCS preserved lung transplant (LTx) recipients at a single, high-volume institution. A total of 22 EVLP and 66 matched SCS LTx recipients were included; SCS grafts were further stratified as either standard-criteria (SCD) or extended-criteria donors (ECD). Median total preservation time was 857, 409, and 438 min for EVLP, SCD, and ECD lungs, respectively (p < .0001). EVLP patients had similar perioperative outcomes and posttransplant survival compared to SCS SCD and ECD recipients. Excluding device-specific costs, total direct variable costs were similar among EVLP, SCD, and ECD recipients (median $200,404, vs. $154,709 vs. $168,334, p =  .11). The median direct contribution margin was positive for EVLP recipients, and similar to that for SCD and ECD graft recipients (all p > .99). These findings demonstrate that the use of EVLP was profitable at an institutional level; however, further investigation is needed to better understand the financial implications of EVLP in facilitating donor pool expansion in an era of broader lung sharing.


Subject(s)
Lung Transplantation , Organ Preservation , Costs and Cost Analysis , Humans , Lung , Lung Transplantation/methods , Organ Preservation/methods , Perfusion/methods , Tissue Donors
4.
ACS Chem Neurosci ; 10(7): 3154-3166, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31140276

ABSTRACT

Serotonin (5-hydroxytryptamine; 5-HT) coordinates behavioral responses to stress through a variety of presynaptic and postsynaptic receptors distributed across functionally diverse neuronal networks in the central nervous system. Efferent 5-HT projections from the dorsal raphe nucleus (DRN) to the bed nucleus of the stria terminalis (BNST) are generally thought to enhance anxiety and aversive learning by activating 5-HT2C receptor (5-HT2CR) signaling in the BNST, although an opposing role for postsynaptic 5-HT1A receptors has recently been suggested. In the present study, we sought to delineate a role for postsynaptic 5-HT1A receptors in the BNST in aversive behaviors using a conditional knockdown of the 5-HT1A receptor. Both males and females were tested to dissect out sex-specific effects. We found that male mice have significantly reduced fear memory recall relative to female mice and inactivation of 5-HT1A receptor in the BNST increases contextual fear conditioning in male mice so that they resemble the females. This coincided with an increase in neuronal excitability in males, suggesting that 5-HT1A receptor deletion may enhance contextual fear recall by disinhibiting fear memory circuits in the BNST. Interestingly, 5-HT1A receptor knockdown did not significantly alter anxiety-like behavior in male or female mice, which is in agreement with previous findings that anxiety and fear are modulated by dissociable circuits in the BNST. Overall, these results suggest that BNST 5-HT1A receptors do not significantly alter behavior under basal conditions, but can act as a molecular brake that buffer against excessive activation of aversive circuits in more threatening contexts.


Subject(s)
Anxiety/metabolism , Fear/physiology , Neurons/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Septal Nuclei/metabolism , Animals , Behavior, Animal/physiology , Feeding Behavior/physiology , Female , Gene Knockdown Techniques , Male , Mice , Mice, Transgenic , Motor Activity/physiology , Receptor, Serotonin, 5-HT1A/genetics , Sex Factors
5.
Neuron ; 102(5): 1037-1052.e7, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31029403

ABSTRACT

Food palatability is one of many factors that drives food consumption, and the hedonic drive to feed is a key contributor to obesity and binge eating. In this study, we identified a population of prepronociceptin-expressing cells in the central amygdala (PnocCeA) that are activated by palatable food consumption. Ablation or chemogenetic inhibition of these cells reduces palatable food consumption. Additionally, ablation of PnocCeA cells reduces high-fat-diet-driven increases in bodyweight and adiposity. PnocCeA neurons project to the ventral bed nucleus of the stria terminalis (vBNST), parabrachial nucleus (PBN), and nucleus of the solitary tract (NTS), and activation of cell bodies in the central amygdala (CeA) or axons in the vBNST, PBN, and NTS produces reward behavior but did not promote feeding of palatable food. These data suggest that the PnocCeA network is necessary for promoting the reinforcing and rewarding properties of palatable food, but activation of this network itself is not sufficient to promote feeding.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Feeding Behavior/physiology , Neurons/metabolism , Protein Precursors/metabolism , Receptors, Opioid/metabolism , Reward , Adiposity , Animals , Body Weight , Central Amygdaloid Nucleus/physiology , Diet, High-Fat , Mice , Neural Pathways , Neurons/physiology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Patch-Clamp Techniques , Protein Precursors/genetics , Receptors, Opioid/genetics , Septal Nuclei/metabolism , Septal Nuclei/physiology , Solitary Nucleus/metabolism , Solitary Nucleus/physiology
6.
Prostate ; 76(1): 97-113, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26417683

ABSTRACT

BACKGROUND: Previous results from our lab indicate a tumor suppressor role for the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) in prostate cancer (PCa). Here, we further characterize this role and uncover new functions for TMEFF2 in cancer and adult prostate regeneration. METHODS: The role of TMEFF2 was examined in PCa cells using Matrigel(TM) cultures and allograft models of PCa cells. In addition, we developed a transgenic mouse model that expresses TMEFF2 from a prostate specific promoter. Anatomical, histological, and metabolic characterizations of the transgenic mouse prostate were conducted. The effect of TMEFF2 in prostate regeneration was studied by analyzing branching morphogenesis in the TMEFF2-expressing mouse lobes and alterations in branching morphogenesis were correlated with the metabolomic profiles of the mouse lobes. The role of TMEFF2 in prostate tumorigenesis in whole animals was investigated by crossing the TMEFF2 transgenic mice with the TRAMP mouse model of PCa and analyzing the histopathological changes in the progeny. RESULTS: Ectopic expression of TMEFF2 impairs growth of PCa cells in Matrigel or allograft models. Surprisingly, while TMEFF2 expression in the TRAMP mouse did not have a significant effect on the glandular prostate epithelial lesions, the double TRAMP/TMEFF2 transgenic mice displayed an increased incidence of neuroendocrine type tumors. In addition, TMEFF2 promoted increased branching specifically in the dorsal lobe of the prostate suggesting a potential role in developmental processes. These results correlated with data indicating an alteration in the metabolic profile of the dorsal lobe of the transgenic TMEFF2 mice. CONCLUSIONS: Collectively, our results confirm the tumor suppressor role of TMEFF2 and suggest that ectopic expression of TMEFF2 in mouse prostate leads to additional lobe-specific effects in prostate regeneration and tumorigenesis. This points to a complex and multifunctional role for TMEFF2 during PCa progression.


Subject(s)
Adenocarcinoma , Carcinogenesis/metabolism , Membrane Proteins/metabolism , Neuroendocrine Tumors , Prostate , Prostatic Neoplasms , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neoplasm Transplantation/pathology , Neoplasm Transplantation/physiology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prostate/pathology , Prostate/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Regeneration , Tumor Cells, Cultured
7.
Biochim Biophys Acta ; 1843(6): 1216-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24632071

ABSTRACT

Cell adhesion and migration play important roles in physiological and pathological states, including embryonic development and cancer invasion and metastasis. The type I transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is expressed mainly in brain and prostate and its expression is deregulated in prostate cancer. We have previously shown that TMEFF2 can function as a tumor suppressor by inhibiting cell migration and invasion of prostate cells. However, the molecular mechanisms involved in this inhibition are not clear. In this study we demonstrate that TMEFF2 affects cell adhesion and migration of prostate cancer cells and that this effect correlates with changes in integrin expression and RhoA activation. Deletion of a 13 basic-rich amino acid region in the cytoplasmic domain of TMEFF2 prevented these effects. Overexpression of TMEFF2 reduced cell attachment and migration on vitronectin and caused a concomitant decrease in RhoA activation, stress fiber formation and expression of αv, ß1 and ß3 integrin subunits. Conversely, TMEFF2 interference in 22Rv1 prostate cancer cells resulted in an increased integrin expression. Results obtained with a double TRAMP/TMEFF2 transgenic mouse also indicated that TMEFF2 expression reduced integrin expression in the mouse prostate. In summary, the data presented here indicate an important role of TMEFF2 in regulating cell adhesion and migration that involves integrin signaling and is mediated by its cytoplasmic domain.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Integrin alphaV/metabolism , Integrin beta3/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cell Shape , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Focal Adhesions , Humans , Integrin alphaV/genetics , Integrin beta3/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...