LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 62

Search options

  1. Article ; Online: Evaluation of long-chain fatty acid respiration in neonatal mouse cardiomyocytes using SeaHorse instrument.

    Angelini, Aude / Pi, Xinchun / Xie, Liang

    STAR protocols

    2022  Volume 3, Issue 2, Page(s) 101392

    Abstract: Metabolic switches play a critical role in the pathophysiology of cardiac diseases, including heart failure. Here, we describe an assay for long-chain fatty acid oxidation in neonatal mouse cardiomyocytes by using a SeaHorse Flux Analyzer (Agilent). This ...

    Abstract Metabolic switches play a critical role in the pathophysiology of cardiac diseases, including heart failure. Here, we describe an assay for long-chain fatty acid oxidation in neonatal mouse cardiomyocytes by using a SeaHorse Flux Analyzer (Agilent). This protocol is a simplified but robust adaptation of the standard protocol that enables metabolic measurements in cells isolated from transgenic mouse models, which can be timesaving and informative. Cell isolation and culture represent a critical point that may require bench optimization. For complete details on the use and execution of this protocol, please refer to Angelini et al. (2021).
    MeSH term(s) Animals ; Animals, Newborn ; Fatty Acids ; Mice ; Myocytes, Cardiac ; Respiration ; Smegmamorpha
    Chemical Substances Fatty Acids
    Language English
    Publishing date 2022-05-17
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ISSN 2666-1667
    ISSN (online) 2666-1667
    DOI 10.1016/j.xpro.2022.101392
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Evaluation of long-chain fatty acid respiration in neonatal mouse cardiomyocytes using SeaHorse instrument

    Aude Angelini / Xinchun Pi / Liang Xie

    STAR Protocols, Vol 3, Iss 2, Pp 101392- (2022)

    2022  

    Abstract: Summary: Metabolic switches play a critical role in the pathophysiology of cardiac diseases, including heart failure. Here, we describe an assay for long-chain fatty acid oxidation in neonatal mouse cardiomyocytes by using a SeaHorse Flux Analyzer ( ... ...

    Abstract Summary: Metabolic switches play a critical role in the pathophysiology of cardiac diseases, including heart failure. Here, we describe an assay for long-chain fatty acid oxidation in neonatal mouse cardiomyocytes by using a SeaHorse Flux Analyzer (Agilent). This protocol is a simplified but robust adaptation of the standard protocol that enables metabolic measurements in cells isolated from transgenic mouse models, which can be timesaving and informative. Cell isolation and culture represent a critical point that may require bench optimization.For complete details on the use and execution of this protocol, please refer to Angelini et al. (2021). : Publisher's note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
    Keywords Cell Biology ; Cell culture ; Cell isolation ; Metabolism ; Protein Biochemistry ; Science (General) ; Q1-390
    Subject code 571
    Language English
    Publishing date 2022-06-01T00:00:00Z
    Publisher Elsevier
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  3. Article ; Online: CRAT links cholesterol metabolism to innate immune responses in the heart.

    Mao, Hua / Angelini, Aude / Li, Shengyu / Wang, Guangyu / Li, Luge / Patterson, Cam / Pi, Xinchun / Xie, Liang

    Nature metabolism

    2023  Volume 5, Issue 8, Page(s) 1382–1394

    Abstract: Chronic inflammation is associated with increased risk and poor prognosis of heart failure; however, the precise mechanism that provokes sustained inflammation in the failing heart remains elusive. Here we report that depletion of carnitine ... ...

    Abstract Chronic inflammation is associated with increased risk and poor prognosis of heart failure; however, the precise mechanism that provokes sustained inflammation in the failing heart remains elusive. Here we report that depletion of carnitine acetyltransferase (CRAT) promotes cholesterol catabolism through bile acid synthesis pathway in cardiomyocytes. Intracellular accumulation of bile acid or intermediate, 7α-hydroxyl-3-oxo-4-cholestenoic acid, induces mitochondrial DNA stress and triggers cGAS-STING-dependent type I interferon responses. Furthermore, type I interferon responses elicited by CRAT deficiency substantially increase AIM2 expression and AIM2-dependent inflammasome activation. Genetic deletion of cardiomyocyte CRAT in mice of both sexes results in myocardial inflammation and dilated cardiomyopathy, which can be reversed by combined depletion of caspase-1, cGAS or AIM2. Collectively, we identify a mechanism by which cardiac energy metabolism, cholesterol homeostasis and cardiomyocyte-intrinsic innate immune responses are interconnected via a CRAT-mediated bile acid synthesis pathway, which contributes to chronic myocardial inflammation and heart failure progression.
    MeSH term(s) Animals ; Female ; Male ; Mice ; Carnitine O-Acetyltransferase/genetics ; Carnitine O-Acetyltransferase/metabolism ; Cholesterol ; Heart Failure ; Immunity, Innate ; Inflammation ; Interferon Type I ; Nucleotidyltransferases/metabolism
    Chemical Substances Carnitine O-Acetyltransferase (EC 2.3.1.7) ; Cholesterol (97C5T2UQ7J) ; Interferon Type I ; Nucleotidyltransferases (EC 2.7.7.-)
    Language English
    Publishing date 2023-07-13
    Publishing country Germany
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ISSN 2522-5812
    ISSN (online) 2522-5812
    DOI 10.1038/s42255-023-00844-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Emerging Roles of Vascular Endothelium in Metabolic Homeostasis.

    Pi, Xinchun / Xie, Liang / Patterson, Cam

    Circulation research

    2018  Volume 123, Issue 4, Page(s) 477–494

    Abstract: Our understanding of the role of the vascular endothelium has evolved over the past 2 decades, with the recognition that it is a dynamically regulated organ and that it plays a nodal role in a variety of physiological and pathological processes. ... ...

    Abstract Our understanding of the role of the vascular endothelium has evolved over the past 2 decades, with the recognition that it is a dynamically regulated organ and that it plays a nodal role in a variety of physiological and pathological processes. Endothelial cells (ECs) are not only a barrier between the circulation and peripheral tissues, but also actively regulate vascular tone, blood flow, and platelet function. Dysregulation of ECs contributes to pathological conditions such as vascular inflammation, atherosclerosis, hypertension, cardiomyopathy, retinopathy, neuropathy, and cancer. The close anatomic relationship between vascular endothelium and highly vascularized metabolic organs/tissues suggests that the crosstalk between ECs and these organs is vital for both vascular and metabolic homeostasis. Numerous reports support that hyperlipidemia, hyperglycemia, and other metabolic stresses result in endothelial dysfunction and vascular complications. However, how ECs may regulate metabolic homeostasis remains poorly understood. Emerging data suggest that the vascular endothelium plays an unexpected role in the regulation of metabolic homeostasis and that endothelial dysregulation directly contributes to the development of metabolic disorders. Here, we review recent studies about the pivotal role of ECs in glucose and lipid homeostasis. In particular, we introduce the concept that the endothelium adjusts its barrier function to control the transendothelial transport of fatty acids, lipoproteins, LPLs (lipoprotein lipases), glucose, and insulin. In addition, we summarize reports that ECs communicate with metabolic cells through EC-secreted factors and we discuss how endothelial dysregulation contributes directly to the development of obesity, insulin resistance, dyslipidemia, diabetes mellitus, cognitive defects, and fatty liver disease.
    MeSH term(s) Animals ; Endothelium, Vascular/metabolism ; Energy Metabolism ; Homeostasis ; Humans ; Metabolic Diseases/etiology
    Language English
    Publishing date 2018-10-24
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Review
    ZDB-ID 80100-8
    ISSN 1524-4571 ; 0009-7330 ; 0931-6876
    ISSN (online) 1524-4571
    ISSN 0009-7330 ; 0931-6876
    DOI 10.1161/CIRCRESAHA.118.313237
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Prolyl Hydroxylase Domain-2 Protein Regulates Lipopolysaccharide-Induced Vascular Inflammation.

    Fan, Qiying / Mao, Hua / Xie, Liang / Pi, Xinchun

    The American journal of pathology

    2018  Volume 189, Issue 1, Page(s) 200–213

    Abstract: Acute lung injury and its more severe form, acute respiratory distress syndrome, are life-threatening respiratory disorders. Overwhelming pulmonary inflammation and endothelium disruption are commonly observed. Endothelial cells (ECs) are well recognized ...

    Abstract Acute lung injury and its more severe form, acute respiratory distress syndrome, are life-threatening respiratory disorders. Overwhelming pulmonary inflammation and endothelium disruption are commonly observed. Endothelial cells (ECs) are well recognized as key regulators in leukocyte adhesion and migration in response to bacterial infection. Prolyl hydroxylase domain (PHD)-2 protein, a major PHD in ECs, plays a critical role in intracellular oxygen homeostasis, angiogenesis, and pulmonary hypertension. However, its role in endothelial inflammatory response is unclear. We investigated the role of PHD2 in ECs during endotoxin-induced lung inflammatory responses with EC-specific PHD2 inducible knockout mice. On lipopolysaccharide challenge, PHD2 depletion in ECs attenuates lipopolysaccharide-induced increases of lung vascular permeability, edema, and inflammatory cell infiltration. Moreover, EC-specific PHD2 inducible knockout mice exhibit improved adherens junction integrity and endothelial barrier function. Mechanistically, PHD2 knockdown induces vascular endothelial cadherin in mouse lung microvascular primary endothelial cells. Moreover, PHD2 knockdown can increase hypoxia-inducible factor/vascular endothelial protein tyrosine phosphatase signaling and reactive oxygen species-dependent p38 activation, leading to the induction of vascular endothelial cadherin. Data indicate that PHD2 depletion prevents the formation of leaky vessels and edema by regulating endothelial barrier function. It provides direct in vivo evidence to suggest that PHD2 plays a pivotal role in vascular inflammation. The inhibition of endothelial PHD2 activity may be a new therapeutic strategy for acute inflammatory diseases.
    MeSH term(s) Acute Lung Injury/chemically induced ; Acute Lung Injury/genetics ; Acute Lung Injury/immunology ; Acute Lung Injury/pathology ; Alveolar Epithelial Cells/immunology ; Alveolar Epithelial Cells/pathology ; Animals ; Cadherins/genetics ; Cadherins/immunology ; Capillary Permeability/drug effects ; Capillary Permeability/genetics ; Capillary Permeability/immunology ; Cell Adhesion/drug effects ; Cell Adhesion/genetics ; Cell Adhesion/immunology ; Cell Movement/drug effects ; Cell Movement/genetics ; Cell Movement/immunology ; Endothelial Cells/immunology ; Endothelial Cells/pathology ; Endothelium, Vascular/immunology ; Endothelium, Vascular/pathology ; Humans ; Hypoxia-Inducible Factor-Proline Dioxygenases/genetics ; Hypoxia-Inducible Factor-Proline Dioxygenases/immunology ; Inflammation/chemically induced ; Inflammation/genetics ; Inflammation/immunology ; Inflammation/pathology ; Leukocytes/immunology ; Leukocytes/pathology ; Lipopolysaccharides/toxicity ; MAP Kinase Signaling System/drug effects ; MAP Kinase Signaling System/genetics ; MAP Kinase Signaling System/immunology ; Male ; Mice ; Mice, Transgenic ; Protein Tyrosine Phosphatases/genetics ; Protein Tyrosine Phosphatases/immunology ; United States ; Vasculitis/chemically induced ; Vasculitis/genetics ; Vasculitis/immunology ; Vasculitis/pathology ; p38 Mitogen-Activated Protein Kinases/genetics ; p38 Mitogen-Activated Protein Kinases/immunology
    Chemical Substances Cadherins ; Cdh1 protein, mouse ; Lipopolysaccharides ; Egln1 protein, mouse (EC 1.14.11.29) ; Hypoxia-Inducible Factor-Proline Dioxygenases (EC 1.14.11.29) ; p38 Mitogen-Activated Protein Kinases (EC 2.7.11.24) ; Protein Tyrosine Phosphatases (EC 3.1.3.48)
    Language English
    Publishing date 2018-10-17
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2943-9
    ISSN 1525-2191 ; 0002-9440
    ISSN (online) 1525-2191
    ISSN 0002-9440
    DOI 10.1016/j.ajpath.2018.09.012
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article: Dioxygen and Metabolism; Dangerous Liaisons in Cardiac Function and Disease.

    Angelini, Aude / Pi, Xinchun / Xie, Liang

    Frontiers in physiology

    2017  Volume 8, Page(s) 1044

    Abstract: The heart must consume a significant amount of energy to sustain its contractile activity. Although the fuel demands are huge, the stock remains very low. Thus, in order to supply its daily needs, the heart must have amazing adaptive abilities, which are ...

    Abstract The heart must consume a significant amount of energy to sustain its contractile activity. Although the fuel demands are huge, the stock remains very low. Thus, in order to supply its daily needs, the heart must have amazing adaptive abilities, which are dependent on dioxygen availability. However, in myriad cardiovascular diseases, "fuel" depletion and hypoxia are common features, leading cardiomyocytes to favor low-dioxygen-consuming glycolysis rather than oxidation of fatty acids. This metabolic switch makes it challenging to distinguish causes from consequences in cardiac pathologies. Finally, despite the progress achieved in the past few decades, medical treatments have not improved substantially, either. In such a situation, it seems clear that much remains to be learned about cardiac diseases. Therefore, in this review, we will discuss how reconciling dioxygen availability and cardiac metabolic adaptations may contribute to develop full and innovative strategies from bench to bedside.
    Language English
    Publishing date 2017-12-12
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2564217-0
    ISSN 1664-042X
    ISSN 1664-042X
    DOI 10.3389/fphys.2017.01044
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article: Low-Density Lipoprotein Receptor-Related Protein-1 Signaling in Angiogenesis.

    Mao, Hua / Xie, Liang / Pi, Xinchun

    Frontiers in cardiovascular medicine

    2017  Volume 4, Page(s) 34

    Abstract: Low-density lipoprotein receptor-related protein-1 (LRP1) plays multifunctional roles in lipid homeostasis, signaling transduction, and endocytosis. It has been recognized as an endocytic receptor for many ligands and is involved in the signaling ... ...

    Abstract Low-density lipoprotein receptor-related protein-1 (LRP1) plays multifunctional roles in lipid homeostasis, signaling transduction, and endocytosis. It has been recognized as an endocytic receptor for many ligands and is involved in the signaling pathways of many growth factors or cytokines. Dysregulation of LRP1-dependent signaling events contributes to the development of pathophysiologic processes such as Alzheimer's disease, atherosclerosis, inflammation, and coagulation. Interestingly, recent studies have linked LRP1 with endothelial function and angiogenesis, which has been underappreciated for a long time. During zebrafish embryonic development, LRP1 is required for the formation of vascular network, especially for the venous development. LRP1 depletion in the mouse embryo proper leads to angiogenic defects and disruption of endothelial integrity. Moreover, in a mouse oxygen-induced retinopathy model, specific depletion of LRP1 in endothelial cells results in abnormal development of neovessels. These loss-of-function studies suggest that LRP1 plays a pivotal role in angiogenesis. The review addresses the recent advances in the roles of LRP1-dependent signaling during angiogenesis.
    Language English
    Publishing date 2017-05-22
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2781496-8
    ISSN 2297-055X
    ISSN 2297-055X
    DOI 10.3389/fcvm.2017.00034
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: PHDs/CPT1B/VDAC1 axis regulates long-chain fatty acid oxidation in cardiomyocytes

    Aude Angelini / Pradip K. Saha / Antrix Jain / Sung Yun Jung / Randall L. Mynatt / Xinchun Pi / Liang Xie

    Cell Reports, Vol 37, Iss 1, Pp 109767- (2021)

    2021  

    Abstract: Summary: Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported ... ...

    Abstract Summary: Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ischemic or late-stage failing hearts. The mechanism regulating this metabolic switch remains poorly understood. Here, we report that loss of PHD2/3, the cellular oxygen sensors, blocks LCFA mitochondria uptake and β-oxidation in cardiomyocytes. In high-fat-fed mice, PHD2/3 deficiency improves glucose metabolism but exacerbates the cardiac defects. Mechanistically, we find that PHD2/3 bind to CPT1B, a key enzyme of mitochondrial LCFA uptake, promoting CPT1B-P295 hydroxylation. Further, we show that CPT1B-P295 hydroxylation is indispensable for its interaction with VDAC1 and LCFA β-oxidation. Finally, we demonstrate that a CPT1B-P295A mutant constitutively binds to VDAC1 and rescues LCFA metabolism in PHD2/3-deficient cardiomyocytes. Together, our data identify an oxygen-sensitive regulatory axis involved in cardiac metabolism.
    Keywords cardiac metabolism switch ; carnitine O-palmitoyltransferase 1b ; myocardial infarction ; heart failure ; hypoxia ; long-chain fatty acid ; Biology (General) ; QH301-705.5
    Language English
    Publishing date 2021-10-01T00:00:00Z
    Publisher Elsevier
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  9. Article ; Online: PHDs/CPT1B/VDAC1 axis regulates long-chain fatty acid oxidation in cardiomyocytes.

    Angelini, Aude / Saha, Pradip K / Jain, Antrix / Jung, Sung Yun / Mynatt, Randall L / Pi, Xinchun / Xie, Liang

    Cell reports

    2021  Volume 37, Issue 1, Page(s) 109767

    Abstract: Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ... ...

    Abstract Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ischemic or late-stage failing hearts. The mechanism regulating this metabolic switch remains poorly understood. Here, we report that loss of PHD2/3, the cellular oxygen sensors, blocks LCFA mitochondria uptake and β-oxidation in cardiomyocytes. In high-fat-fed mice, PHD2/3 deficiency improves glucose metabolism but exacerbates the cardiac defects. Mechanistically, we find that PHD2/3 bind to CPT1B, a key enzyme of mitochondrial LCFA uptake, promoting CPT1B-P295 hydroxylation. Further, we show that CPT1B-P295 hydroxylation is indispensable for its interaction with VDAC1 and LCFA β-oxidation. Finally, we demonstrate that a CPT1B-P295A mutant constitutively binds to VDAC1 and rescues LCFA metabolism in PHD2/3-deficient cardiomyocytes. Together, our data identify an oxygen-sensitive regulatory axis involved in cardiac metabolism.
    MeSH term(s) Animals ; Carnitine/metabolism ; Carnitine O-Palmitoyltransferase/deficiency ; Carnitine O-Palmitoyltransferase/genetics ; Carnitine O-Palmitoyltransferase/metabolism ; Diet, High-Fat ; Fatty Acids/chemistry ; Fatty Acids/metabolism ; Glucose/metabolism ; Hydroxylation ; Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency ; Hypoxia-Inducible Factor-Proline Dioxygenases/genetics ; Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism ; Lipid Peroxidation ; Mice ; Mice, Inbred C57BL ; Mice, Knockout ; Mitochondria/metabolism ; Mutagenesis, Site-Directed ; Myocytes, Cardiac/cytology ; Myocytes, Cardiac/metabolism ; Procollagen-Proline Dioxygenase/deficiency ; Procollagen-Proline Dioxygenase/genetics ; Procollagen-Proline Dioxygenase/metabolism ; Protein Binding ; Voltage-Dependent Anion Channel 1/genetics ; Voltage-Dependent Anion Channel 1/metabolism
    Chemical Substances Fatty Acids ; Vdac1 protein, mouse ; PHD3 protein, mouse (EC 1.14.11.2) ; Procollagen-Proline Dioxygenase (EC 1.14.11.2) ; Egln1 protein, mouse (EC 1.14.11.29) ; Hypoxia-Inducible Factor-Proline Dioxygenases (EC 1.14.11.29) ; Voltage-Dependent Anion Channel 1 (EC 1.6.-) ; CPT1B protein, mouse (EC 2.3.1.21) ; Carnitine O-Palmitoyltransferase (EC 2.3.1.21) ; Glucose (IY9XDZ35W2) ; Carnitine (S7UI8SM58A)
    Language English
    Publishing date 2021-10-05
    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.2021.109767
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Safety and efficiency of stem cell therapy for COVID-19: a systematic review and meta-analysis.

    Zhang, Minghe / Yan, Xinchun / Shi, Minghui / Li, Ruihang / Pi, Ziwei / Ren, Xiangying / Wang, Yongbo / Yan, Siyu / Wang, Yunyun / Jin, Yinghui / Wang, Xinghuan

    Global health research and policy

    2022  Volume 7, Issue 1, Page(s) 19

    Abstract: Background: With the COVID-19 pandemic continuing, various treatments have become widely practiced. Stem cells have a wide range of applications in the treatment of lung diseases and have therefore been experimentally used to treat patients with COVID- ... ...

    Abstract Background: With the COVID-19 pandemic continuing, various treatments have become widely practiced. Stem cells have a wide range of applications in the treatment of lung diseases and have therefore been experimentally used to treat patients with COVID-19, but whether the expanded use of stem cells is safe and reliable still lacks enough evidence. To address this issue, we systematically reviewed the safety and efficiency of stem cell therapy in COVID-19 cases.
    Methods: We searched PubMed, Embase, Web of Science, The Cochrane Library, CNKI, WanFang, VIP and SinoMed up to January 18, 2022. The included studies were assessed using the Risk-of-bias tool 1.0 and MINORS instrument. The adverse events, mortality, length of hospital day and laboratory parameters were analyzed by meta-analysis. We adhered to PRISMA reporting guideline.
    Results: We have included 17 studies meeting the inclusion data. There were no significant differences in AEs (OR = 0·39, 95% CI = 0·12 to 1·33, P = 0·13, I
    Conclusions: The present study shows that stem cell therapy for COVID-19 has a remarkable effect on efficiency without increasing risks of adverse events and length of hospital stay. It is potentially necessary to establish the criteria for COVID-19 for stem cell therapy.
    MeSH term(s) Bias ; COVID-19/therapy ; Cell- and Tissue-Based Therapy ; Humans ; Pandemics ; SARS-CoV-2
    Language English
    Publishing date 2022-06-23
    Publishing country England
    Document type Journal Article ; Meta-Analysis ; Review ; Systematic Review ; Research Support, Non-U.S. Gov't
    ISSN 2397-0642
    ISSN (online) 2397-0642
    DOI 10.1186/s41256-022-00251-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top