LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 10

Search options

  1. Article: A novel cross-institutional college internship program to train future diverse leaders in clinical research with data-driven approaches to assess impact.

    Derk, Julia / Dzirasa, Kafui / Locklear, Tracie

    Frontiers in pharmacology

    2023  Volume 14, Page(s) 1294535

    Abstract: The field of Clinical Research, like many other scientific disciplines, has struggled to recruit and retain talented researchers from diverse communities. While there is a strong history of documenting the problem, having a diverse and inclusive ... ...

    Abstract The field of Clinical Research, like many other scientific disciplines, has struggled to recruit and retain talented researchers from diverse communities. While there is a strong history of documenting the problem, having a diverse and inclusive workforce is hindered by the lack of data-driven approaches, cross-institutional partnerships, access to mentors, and positive immersive experiences for people from underrepresented groups. Here, we describe a novel initiative for North Carolina Central University Clinical Research Sciences Program (NCCU-CRSP) student interns to partner with Duke University to have immersive clinical and pre-clinical research training in a 15-week internship as the culminating experience towards their degree for a Bachelor of Science in Clinical Research. The goals of the internship are: 1) to give hands-on training to enhance the impact of classroom-based learning, 2) broaden their understanding of the wide swath of positions available to them, 3) promote their sense of self-efficacy, confidence, science identity, research identity, and connections to the pre-clinical and clinical community, and 4) prepare them to be workforce ready upon graduating. The students dedicate 75% of their time to clinical research with Duke University at Pickett Road and 25% to pre-clinical research in the Collective for Psychiatric Neuroengineering in the Duke Psychiatry Department of the School of Medicine. They will also receive eight 1-h professional development training sessions from the Duke-NCCU Clinical and Translational Science Initiative's Workforce Development Team and five 1-h sessions based on the Entering Research Curriculum developed by the Center for the Improvement of Mentored Experiences in Research (CIMER). Finally, they will be brought in as a cohort and coached on peer mentoring and mutual support frameworks to enhance their sense of community. These student-interns will perform pre- and post-internship self-assessment surveys to quantify their self-efficacy, feelings of belonging, access to research opportunities and mentors, and to give details of their future education and career goals. We will evaluate the impact of the internship using validated tools and apply these findings for future optimization of program design and tactical advice for other programs with shared missions. Furthermore, we will email them on an annual basis with follow-up surveys to assess the longitudinal impact of this internship program, their educational experiences at NCCU, what job titles they hold, how prepared they feel for their roles, and what they hope their future career trajectory will be. Collectively, these approaches will apply theoretical frameworks developed by social and cognitive psychology, vocational theory, and educational research to clinical research training with the goals of recruiting and training talented and diverse leaders within clinical research. We hope that by evaluating our successes, failures, strengths, and liabilities through empirically derived evidence we will also inspire future studies that use data-driven approaches to elevate our approaches as we work together to train and recruit talented researchers from diverse communities into our scientific enterprise and to launch them with more in-depth experiential learning that will empower them to succeed.
    Language English
    Publishing date 2023-12-06
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2587355-6
    ISSN 1663-9812
    ISSN 1663-9812
    DOI 10.3389/fphar.2023.1294535
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Corrigendum: Living on the Edge of the CNS: Meninges Cell Diversity in Health and Disease.

    Derk, Julia / Jones, Hannah E / Como, Christina / Pawlikowski, Bradley / Siegenthaler, Julie A

    Frontiers in cellular neuroscience

    2021  Volume 15, Page(s) 761506

    Abstract: This corrects the article DOI: 10.3389/fncel.2021.703944.]. ...

    Abstract [This corrects the article DOI: 10.3389/fncel.2021.703944.].
    Language English
    Publishing date 2021-10-08
    Publishing country Switzerland
    Document type Published Erratum
    ZDB-ID 2452963-1
    ISSN 1662-5102
    ISSN 1662-5102
    DOI 10.3389/fncel.2021.761506
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: Living on the Edge of the CNS: Meninges Cell Diversity in Health and Disease.

    Derk, Julia / Jones, Hannah E / Como, Christina / Pawlikowski, Bradley / Siegenthaler, Julie A

    Frontiers in cellular neuroscience

    2021  Volume 15, Page(s) 703944

    Abstract: The meninges are the fibrous covering of the central nervous system (CNS) which contain vastly heterogeneous cell types within its three layers (dura, arachnoid, and pia). The dural compartment of the meninges, closest to the skull, is predominantly ... ...

    Abstract The meninges are the fibrous covering of the central nervous system (CNS) which contain vastly heterogeneous cell types within its three layers (dura, arachnoid, and pia). The dural compartment of the meninges, closest to the skull, is predominantly composed of fibroblasts, but also includes fenestrated blood vasculature, an elaborate lymphatic system, as well as immune cells which are distinct from the CNS. Segregating the outer and inner meningeal compartments is the epithelial-like arachnoid barrier cells, connected by tight and adherens junctions, which regulate the movement of pathogens, molecules, and cells into and out of the cerebral spinal fluid (CSF) and brain parenchyma. Most proximate to the brain is the collagen and basement membrane-rich pia matter that abuts the glial limitans and has recently be shown to have regional heterogeneity within the developing mouse brain. While the meninges were historically seen as a purely structural support for the CNS and protection from trauma, the emerging view of the meninges is as an essential interface between the CNS and the periphery, critical to brain development, required for brain homeostasis, and involved in a variety of diseases. In this review, we will summarize what is known regarding the development, specification, and maturation of the meninges during homeostatic conditions and discuss the rapidly emerging evidence that specific meningeal cell compartments play differential and important roles in the pathophysiology of a myriad of diseases including: multiple sclerosis, dementia, stroke, viral/bacterial meningitis, traumatic brain injury, and cancer. We will conclude with a list of major questions and mechanisms that remain unknown, the study of which represent new, future directions for the field of meninges biology.
    Language English
    Publishing date 2021-07-01
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2452963-1
    ISSN 1662-5102
    ISSN 1662-5102
    DOI 10.3389/fncel.2021.703944
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: The receptor for advanced glycation endproducts is a mediator of toxicity by IAPP and other proteotoxic aggregates: Establishing and exploiting common ground for novel amyloidosis therapies.

    Abedini, Andisheh / Derk, Julia / Schmidt, Ann Marie

    Protein science : a publication of the Protein Society

    2018  Volume 27, Issue 7, Page(s) 1166–1180

    Abstract: Proteotoxicity plays a key role in many devastating human disorders, including Alzheimer's, Huntington's and Parkinson's diseases; type 2 diabetes; systemic amyloidosis; and cardiac dysfunction, to name a few. The cellular mechanisms of proteotoxicity in ...

    Abstract Proteotoxicity plays a key role in many devastating human disorders, including Alzheimer's, Huntington's and Parkinson's diseases; type 2 diabetes; systemic amyloidosis; and cardiac dysfunction, to name a few. The cellular mechanisms of proteotoxicity in these disorders have been the focus of considerable research, but their role in prevalent and morbid disorders, such as diabetes, is less appreciated. There is a large body of literature on the impact of glucotoxicity and lipotoxicity on insulin-producing pancreatic β-cells, and there is increasing recognition that proteotoxicty plays a key role. Pancreatic islet amyloidosis by the hormone IAPP, the production of advanced glycation endproducts (AGE), and insulin misprocessing into cytotoxic aggregates are all sources of β-cell proteotoxicity in diabetes. AGE, produced by the reaction of reducing sugars with proteins and lipids are ligands for the receptor for AGE (RAGE), as are the toxic pre-fibrillar aggregates of IAPP produced during amyloid formation. The mechanisms of amyloid formation by IAPP in vivo or in vitro are not well understood, and the cellular mechanisms of IAPP-induced β-cell death are not fully defined. Here, we review recent findings that illuminate the factors and mechanisms involved in β-cell proteotoxicity in diabetes. Together, these new insights have far-reaching implications for the establishment of unifying mechanisms by which pathological amyloidoses imbue their injurious effects in vivo.
    MeSH term(s) Animals ; Cell Survival/drug effects ; Diabetes Mellitus, Type 2/etiology ; Diabetes Mellitus, Type 2/metabolism ; Humans ; Insulin-Secreting Cells/cytology ; Insulin-Secreting Cells/drug effects ; Insulin-Secreting Cells/metabolism ; Islet Amyloid Polypeptide/toxicity ; Receptor for Advanced Glycation End Products/metabolism
    Chemical Substances Islet Amyloid Polypeptide ; Receptor for Advanced Glycation End Products
    Language English
    Publishing date 2018-04-25
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1106283-6
    ISSN 1469-896X ; 0961-8368
    ISSN (online) 1469-896X
    ISSN 0961-8368
    DOI 10.1002/pro.3425
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Formation and function of the meningeal arachnoid barrier around the developing mouse brain.

    Derk, Julia / Como, Christina N / Jones, Hannah E / Joyce, Luke R / Kim, Sol / Spencer, Brady L / Bonney, Stephanie / O'Rourke, Rebecca / Pawlikowski, Brad / Doran, Kelly S / Siegenthaler, Julie A

    Developmental cell

    2023  Volume 58, Issue 8, Page(s) 635–644.e4

    Abstract: The arachnoid barrier, a component of the blood-cerebrospinal fluid barrier (B-CSFB) in the meninges, is composed of epithelial-like, tight-junction-expressing cells. Unlike other central nervous system (CNS) barriers, its' developmental mechanisms and ... ...

    Abstract The arachnoid barrier, a component of the blood-cerebrospinal fluid barrier (B-CSFB) in the meninges, is composed of epithelial-like, tight-junction-expressing cells. Unlike other central nervous system (CNS) barriers, its' developmental mechanisms and timing are largely unknown. Here, we show that mouse arachnoid barrier cell specification requires the repression of Wnt-β-catenin signaling and that constitutively active β-catenin can prevent its formation. We also show that the arachnoid barrier is functional prenatally and, in its absence, a small molecular weight tracer and the bacterium group B Streptococcus can cross into the CNS following peripheral injection. Acquisition of barrier properties prenatally coincides with the junctional localization of Claudin 11, and increased E-cadherin and maturation continues after birth, where postnatal expansion is marked by proliferation and re-organization of junctional domains. This work identifies fundamental mechanisms that drive arachnoid barrier formation, highlights arachnoid barrier fetal functions, and provides novel tools for future studies on CNS barrier development.
    MeSH term(s) Mice ; Animals ; beta Catenin ; Meninges ; Arachnoid ; Blood-Brain Barrier ; Central Nervous System ; Tight Junctions
    Chemical Substances beta Catenin
    Language English
    Publishing date 2023-03-29
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2054967-2
    ISSN 1878-1551 ; 1534-5807
    ISSN (online) 1878-1551
    ISSN 1534-5807
    DOI 10.1016/j.devcel.2023.03.005
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article: The Receptor for Advanced Glycation Endproducts (RAGE) and Mediation of Inflammatory Neurodegeneration.

    Derk, Julia / MacLean, Michael / Juranek, Judyta / Schmidt, Ann Marie

    Journal of Alzheimer's disease & Parkinsonism

    2018  Volume 8, Issue 1

    Language English
    Publishing date 2018-01-24
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2711981-6
    ISSN 2161-0460
    ISSN 2161-0460
    DOI 10.4172/2161-0460.1000421
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: The Receptor for Advanced Glycation End Products (RAGE) and DIAPH1: Implications for vascular and neuroinflammatory dysfunction in disorders of the central nervous system.

    MacLean, Michael / Derk, Julia / Ruiz, Henry H / Juranek, Judyta K / Ramasamy, Ravichandran / Schmidt, Ann Marie

    Neurochemistry international

    2019  Volume 126, Page(s) 154–164

    Abstract: The Receptor for Advanced Glycation End Products (RAGE) is expressed by multiple cell types in the brain and spinal cord that are linked to the pathogenesis of neurovascular and neurodegenerative disorders, including neurons, glia (microglia and ... ...

    Abstract The Receptor for Advanced Glycation End Products (RAGE) is expressed by multiple cell types in the brain and spinal cord that are linked to the pathogenesis of neurovascular and neurodegenerative disorders, including neurons, glia (microglia and astrocytes) and vascular cells (endothelial cells, smooth muscle cells and pericytes). Mounting structural and functional evidence implicates the interaction of the RAGE cytoplasmic domain with the formin, Diaphanous1 (DIAPH1), as the key cytoplasmic hub for RAGE ligand-mediated activation of cellular signaling. In aging and diabetes, the ligands of the receptor abound, both in the central nervous system (CNS) and in the periphery. Such accumulation of RAGE ligands triggers multiple downstream events, including upregulation of RAGE itself. Once set in motion, cell intrinsic and cell-cell communication mechanisms, at least in part via RAGE, trigger dysfunction in the CNS. A key outcome of endothelial dysfunction is reduction in cerebral blood flow and increased permeability of the blood brain barrier, conditions that facilitate entry of activated leukocytes into the CNS, thereby amplifying primary nodes of CNS cellular stress. This contribution details a review of the ligands of RAGE, the mechanisms and consequences of RAGE signal transduction, and cites multiple examples of published work in which RAGE contributes to the pathogenesis of neurovascular perturbation. Insights into potential therapeutic modalities targeting the RAGE signal transduction axis for disorders of CNS vascular dysfunction and neurodegeneration are also discussed.
    MeSH term(s) Animals ; Central Nervous System Diseases/metabolism ; Central Nervous System Diseases/pathology ; Formins/metabolism ; Glycation End Products, Advanced/metabolism ; Humans ; Inflammation Mediators/metabolism ; Neurodegenerative Diseases/metabolism ; Neurodegenerative Diseases/pathology ; Receptor for Advanced Glycation End Products/metabolism ; Vascular Diseases/metabolism ; Vascular Diseases/pathology
    Chemical Substances DIAPH1 protein, human ; Formins ; Glycation End Products, Advanced ; Inflammation Mediators ; Receptor for Advanced Glycation End Products
    Language English
    Publishing date 2019-03-20
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 283190-9
    ISSN 1872-9754 ; 0197-0186
    ISSN (online) 1872-9754
    ISSN 0197-0186
    DOI 10.1016/j.neuint.2019.03.012
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Diaphanous 1 (DIAPH1) is Highly Expressed in the Aged Human Medial Temporal Cortex and Upregulated in Myeloid Cells During Alzheimer's Disease.

    Derk, Julia / Bermudez Hernandez, Keria / Rodriguez, Moises / He, Meilun / Koh, Hyunwook / Abedini, Andisheh / Li, Huilin / Fenyö, David / Schmidt, Ann Marie

    Journal of Alzheimer's disease : JAD

    2018  Volume 64, Issue 3, Page(s) 995–1007

    Abstract: Background: The receptor for advanced glycation end products (RAGE) is linked to cellular stress and inflammation during Alzheimer's disease (AD). RAGE signals through Diaphanous-1 (DIAPH1); however, the expression of DIAPH1 in the healthy and AD human ... ...

    Abstract Background: The receptor for advanced glycation end products (RAGE) is linked to cellular stress and inflammation during Alzheimer's disease (AD). RAGE signals through Diaphanous-1 (DIAPH1); however, the expression of DIAPH1 in the healthy and AD human brain has yet to be methodically addressed.
    Objective: To delineate the cell- and disease-state specific expression of DIAPH1 in the human medial temporal cortex during healthy aging and AD.
    Methods: We used semi-quantitative immunohistochemistry in the human medial temporal cortex paired with widefield and confocal microscopy and automated analyses to determine colocalization and relative expression of DIAPH1 with key cell markers and molecules in the brains of subjects with AD versus age-matched controls.
    Results: We report robust colocalization of DIAPH1 with myeloid cells and increased expression during AD, which strongly correlated to increased neutral lipids and morphology of inflamed myeloid cells. DIAPH1 moderately colocalized with markers of endothelial cells, astrocytes, neurons, and oligodendrocytes.
    Discussion: Our findings localize DIAPH1 particularly to myeloid cells in the CNS, especially in AD in the locations of lipid droplet accumulation, thereby implicating RAGE-DIAPH1 signaling in dysregulated lipid metabolism and morphological changes of inflamed myeloid cells in this disorder.
    MeSH term(s) Adaptor Proteins, Signal Transducing/genetics ; Adaptor Proteins, Signal Transducing/metabolism ; Aged ; Aged, 80 and over ; Aging/pathology ; Alzheimer Disease/genetics ; Alzheimer Disease/pathology ; Animals ; Apolipoproteins E/genetics ; Case-Control Studies ; Claudin-1/metabolism ; Female ; Formins ; Glial Fibrillary Acidic Protein/metabolism ; Humans ; Imaging, Three-Dimensional ; Male ; Mice ; Mice, Inbred C57BL ; Mice, Transgenic ; Microscopy, Confocal ; Microtubule-Associated Proteins/metabolism ; Myeloid Cells/metabolism ; Receptor for Advanced Glycation End Products/metabolism ; Temporal Lobe/metabolism ; Up-Regulation/physiology
    Chemical Substances Adaptor Proteins, Signal Transducing ; Apolipoproteins E ; Claudin-1 ; DIAPH1 protein, human ; Formins ; Glial Fibrillary Acidic Protein ; MAP2 protein, human ; Microtubule-Associated Proteins ; Receptor for Advanced Glycation End Products
    Language English
    Publishing date 2018-06-30
    Publishing country Netherlands
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1440127-7
    ISSN 1875-8908 ; 1387-2877
    ISSN (online) 1875-8908
    ISSN 1387-2877
    DOI 10.3233/JAD-180088
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: RAGE binds preamyloid IAPP intermediates and mediates pancreatic β cell proteotoxicity.

    Abedini, Andisheh / Cao, Ping / Plesner, Annette / Zhang, Jinghua / He, Meilun / Derk, Julia / Patil, Sachi A / Rosario, Rosa / Lonier, Jacqueline / Song, Fei / Koh, Hyunwook / Li, Huilin / Raleigh, Daniel P / Schmidt, Ann Marie

    The Journal of clinical investigation

    2018  Volume 128, Issue 2, Page(s) 682–698

    Abstract: Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in β cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces ...

    Abstract Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in β cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces cellular stress or how IAPP-induced toxicity can be prevented or treated. We recently defined the properties of toxic IAPP species. Here, we have identified a receptor-mediated mechanism of islet amyloidosis-induced proteotoxicity. In human diabetic pancreas and in cellular and mouse models of islet amyloidosis, increased expression of the receptor for advanced glycation endproducts (RAGE) correlated with human IAPP-induced (h-IAPP-induced) β cell and islet inflammation, toxicity, and apoptosis. RAGE selectively bound toxic intermediates, but not nontoxic forms of h-IAPP, including amyloid fibrils. The isolated extracellular ligand-binding domains of soluble RAGE (sRAGE) blocked both h-IAPP toxicity and amyloid formation. Inhibition of the interaction between h-IAPP and RAGE by sRAGE, RAGE-blocking antibodies, or genetic RAGE deletion protected pancreatic islets, β cells, and smooth muscle cells from h-IAPP-induced inflammation and metabolic dysfunction. sRAGE-treated h-IAPP Tg mice were protected from amyloid deposition, loss of β cell area, β cell inflammation, stress, apoptosis, and glucose intolerance. These findings establish RAGE as a mediator of IAPP-induced toxicity and suggest that targeting the IAPP/RAGE axis is a potential strategy to mitigate this source of β cell dysfunction in metabolic disease.
    MeSH term(s) Amyloid/metabolism ; Amyloidosis ; Animals ; Apoptosis ; Cell Line ; Diabetes Mellitus, Type 2/metabolism ; Humans ; Inflammation ; Insulin-Secreting Cells/cytology ; Insulinoma/metabolism ; Islet Amyloid Polypeptide/genetics ; Islets of Langerhans/metabolism ; Ligands ; Male ; Mice ; Mice, Inbred C57BL ; Mice, Transgenic ; Myocytes, Smooth Muscle/metabolism ; Pancreas/metabolism ; Protein Folding ; Rats ; Receptor for Advanced Glycation End Products/metabolism ; Up-Regulation
    Chemical Substances AGER protein, human ; Ager protein, mouse ; Ager protein, rat ; Amyloid ; Islet Amyloid Polypeptide ; Ligands ; Receptor for Advanced Glycation End Products
    Language English
    Publishing date 2018-01-16
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 3067-3
    ISSN 1558-8238 ; 0021-9738
    ISSN (online) 1558-8238
    ISSN 0021-9738
    DOI 10.1172/JCI85210
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: A Receptor of the Immunoglobulin Superfamily Regulates Adaptive Thermogenesis.

    Hurtado Del Pozo, Carmen / Ruiz, Henry H / Arivazhagan, Lakshmi / Aranda, Juan Francisco / Shim, Cynthia / Daya, Peter / Derk, Julia / MacLean, Michael / He, Meilun / Frye, Laura / Friedline, Randall H / Noh, Hye Lim / Kim, Jason K / Friedman, Richard A / Ramasamy, Ravichandran / Schmidt, Ann Marie

    Cell reports

    2019  Volume 28, Issue 3, Page(s) 773–791.e7

    Abstract: Exquisite regulation of energy homeostasis protects from nutrient deprivation but causes metabolic dysfunction upon nutrient excess. In human and murine adipose tissue, the accumulation of ligands of the receptor for advanced glycation end products (RAGE) ...

    Abstract Exquisite regulation of energy homeostasis protects from nutrient deprivation but causes metabolic dysfunction upon nutrient excess. In human and murine adipose tissue, the accumulation of ligands of the receptor for advanced glycation end products (RAGE) accompanies obesity, implicating this receptor in energy metabolism. Here, we demonstrate that mice bearing global- or adipocyte-specific deletion of Ager, the gene encoding RAGE, display superior metabolic recovery after fasting, a cold challenge, or high-fat feeding. The RAGE-dependent mechanisms were traced to suppression of protein kinase A (PKA)-mediated phosphorylation of its key targets, hormone-sensitive lipase and p38 mitogen-activated protein kinase, upon β-adrenergic receptor stimulation-processes that dampen the expression and activity of uncoupling protein 1 (UCP1) and thermogenic programs. This work identifies the innate role of RAGE as a key node in the immunometabolic networks that control responses to nutrient supply and cold challenges, and it unveils opportunities to harness energy expenditure in environmental and metabolic stress.
    MeSH term(s) Adipocytes/enzymology ; Adipocytes/metabolism ; Adipose Tissue/enzymology ; Adipose Tissue/metabolism ; Animals ; Cell Line ; Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors ; Cyclic AMP-Dependent Protein Kinases/metabolism ; Energy Metabolism ; Fasting/metabolism ; Fasting/physiology ; Humans ; Lipolysis/genetics ; Lipolysis/physiology ; Male ; Mice ; Mice, Inbred C57BL ; Mice, Knockout ; Obesity/genetics ; Obesity/metabolism ; Phosphorylation ; Receptor for Advanced Glycation End Products/antagonists & inhibitors ; Receptor for Advanced Glycation End Products/metabolism ; Signal Transduction/genetics ; Signal Transduction/physiology ; Thermogenesis/genetics ; Transplantation, Homologous ; Uncoupling Protein 1/genetics ; Uncoupling Protein 1/metabolism ; p38 Mitogen-Activated Protein Kinases/metabolism
    Chemical Substances Receptor for Advanced Glycation End Products ; Ucp1 protein, mouse ; Uncoupling Protein 1 ; Cyclic AMP-Dependent Protein Kinases (EC 2.7.11.11) ; p38 Mitogen-Activated Protein Kinases (EC 2.7.11.24)
    Language English
    Publishing date 2019-08-06
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2019.06.061
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top