1
|
Luo J, Cui Y, Xu L, Zhang J, Chen J, Li X, Zeng B, Deng Z, Shao L. Layered double hydroxides for regenerative nanomedicine and tissue engineering: recent advances and future perspectives. J Nanobiotechnology 2025; 23:370. [PMID: 40405242 PMCID: PMC12096525 DOI: 10.1186/s12951-025-03448-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
With the rapid development of nanotechnology, layered double hydroxides (LDHs) have attracted considerable attention in the biomedical field due to their highly tunable composition and structure, superior biocompatibility, multifunctional bioactivity, and exceptional drug delivery performance. However, a focused and comprehensive review addressing the role of LDHs specifically in tissue regeneration has been lacking. This review aims to fill that gap by providing a systematic and in-depth overview of recent advances in the application of LDHs across various regenerative domains, including bone repair, cartilage reconstruction, angiogenesis, wound healing, and nerve regeneration. Beyond presenting emerging applications, the review places particular emphasis on elucidating the underlying mechanisms through which LDHs exert their therapeutic effects. Although LDHs demonstrate considerable promise in regenerative medicine, their clinical translation remains in its infancy. To address this, we not only provided our insights into the personalized problems that arise in the application of various tissues, but also focused on discussing and prospecting the common challenges in the clinical translation of LDHs. These challenges include optimizing synthesis techniques, enhancing biosafety and stability, improving drug-loading efficiency, designing multifunctional composite materials, and establishing pathways that facilitate the transition from laboratory research to clinical practice.
Collapse
Affiliation(s)
- Junsi Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Yiteng Cui
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Laijun Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Junyi Zhang
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Jinhong Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Xumin Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Bin Zeng
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Zhiyuan Deng
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- School of Stomatology, Changsha Medical University, Changsha, 410219, China.
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Zhu Y, Mehlkop O, Backes H, Cremer AL, Porniece M, Klemm P, Steuernagel L, Chen W, Johnen R, Wunderlich FT, Jais A, Brüning JC. Reduced Notch signaling in hypothalamic endothelial cells mediates obesity-induced alterations in glucose uptake and insulin signaling. Cell Rep 2025; 44:115522. [PMID: 40186867 DOI: 10.1016/j.celrep.2025.115522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/28/2024] [Accepted: 03/14/2025] [Indexed: 04/07/2025] Open
Abstract
Short-term transition to high-fat diet (HFD) feeding causes rapid changes in the molecular architecture of the blood-brain barrier (BBB), BBB permeability, and brain glucose uptake. However, the precise mechanisms responsible for these changes remain elusive. Here, we detect a rapid downregulation of Notch signaling after short-term HFD feeding. Conversely, Notch activation restores HFD-fed mouse serum-induced reduction of Glut1 expression and glycolysis in cultured brain microvascular endothelial cells (BMECs). Selective, inducible expression of the Notch intracellular domain (IC) in BMECs prevents HFD-induced reduction of Glut1 expression and hypothalamic glucose uptake. Caveolin (Cav)-1 expression in BMECs is increased upon short-term HFD feeding. However, NotchICBMECs mice display reduced caveola formation and BBB permeability. This ultimately translates into reduced hypothalamic insulin transport, action, and systemic insulin sensitivity. Collectively, we highlight a critical role of Notch signaling in the pleiotropic effects of short-term dietary transitions on BBB functionality.
Collapse
Affiliation(s)
- Yiyi Zhu
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Oliver Mehlkop
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Heiko Backes
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Marta Porniece
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
| | - Ronja Johnen
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty & Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany.
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Köln, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
3
|
He T, Chen Q, Li H, Mao J, Luo J, Ma D, Yang Z. The potential mechanism of MicroRNA involvement in the regulation of muscle development in weaned piglets by tryptophan and its metabolites. BMC Genomics 2025; 26:330. [PMID: 40169975 PMCID: PMC11963679 DOI: 10.1186/s12864-025-11424-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Muscle development is a key factor influencing the growth performance of piglets. Optimizing this developmental process is crucial for enhancing breeding efficiency and economic profitability. Tryptophan (Trp) is considered one of the key limiting amino acids for weaned piglets, plays an essential role in regulating feed intake, growth, and muscle development. However, the regulatory mechanisms by which Trp and its derivatives influence muscle development in weaned piglets remain unclear. METHODS The aim of this study was to investigate the regulatory pathways and potential mechanisms of Trp and its metabolites on muscle development in weaned piglets. In this study, 10 healthy castrated male piglets, 28 days old and weaned, were selected and randomly assigned to a control group (CON, 0.14% Trp) and a high tryptophan group (HT, 0.35% Trp), with 5 in each group. After a 7-day pre-feeding period, the formal feeding began, and after 28 days, the pigs were slaughtered and the longissimus dorsi muscles was collected for transcriptome sequencing. RESULTS The results indicated that different dietary Trp levels led to the identification of sixteen differentially expressed microRNAs (DE miRNAs) in the longissimus dorsi muscle of the weaned piglets. Target gene functional enrichment analysis showed that these DE miRNAs are involved in muscle cell proliferation, differentiation, protein deposition, and muscle development through multiple biological pathways. Furthermore, we constructed a protein-protein interaction (PPI) network for the target genes, with the enriched core gene cluster functions associated with cellular proliferation, signaling pathways, hormone release, and muscle development. Finally, qRT-PCR validated the reliability and accuracy of the RNA-seq results, revealing a correlation coefficient of 0.97 between the two methods. CONCLUSIONS This study uncovers the potential mechanisms by which miRNAs participate in the regulation of muscle development in weaned piglets mediated by Trp and its metabolites, providing a theoretical basis and practical guidance for optimizing piglet management and health improvement.
Collapse
Affiliation(s)
- Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Qingyun Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Huifeng Li
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Jiani Mao
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Ju Luo
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Dengjun Ma
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
4
|
Ren G, Bhatnagar S, Young ME, Lee T, Kim JA. Endothelial autophagy-related gene 7 contributes to high fat diet-induced obesity. Mol Metab 2025; 93:102099. [PMID: 39832563 PMCID: PMC11802379 DOI: 10.1016/j.molmet.2025.102099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Obesity-associated metabolic dysfunction is a major public health concern worldwide. Endothelial dysfunction is a hallmark of metabolic dysfunction, and endothelial cells affect metabolic functions. Because autophagy-related gene 7 (ATG7) is involved in various cellular physiology, we investigated the roles of endothelial cell-ATG7 (EC-ATG7) on high-fat diet-induced obesity and its related metabolic dysfunction. METHODS We generated an endothelial-specific Atg7 knock-out mouse by breeding Atg7flox/flox mouse with the Chd5-Cre mouse, and investigated the metabolic phenotypes associated with high-fat diet (HFD)-induced obesity. Body weight, food intake, glucose tolerance, insulin sensitivity, and liver fat accumulation were measured in endothelial Atg7 deficient (Atg7ΔEnd) and control mice (Atg7f/f). Adipose tissue inflammation was assessed by measuring the expression of pro-inflammatory genes. Furthermore, we performed indirect calorimetry and examined the insulin signaling pathway molecules. RESULTS We found that deletion of EC-Atg7 ameliorated HFD-induced weight gain, fatty liver, and adipocyte hypertrophy and inflammatory response in adipose tissue, and improved insulin sensitivity without changing glucose tolerance. These metabolic effects seem to be due to the reduced food intake because there were no differences in energy expenditure, energy excretion to feces, and physical activity. Interestingly, the deletion of EC-Atg7 protected from HFD-induced vascular rarefaction, and the knock-down of Atg7 in endothelial cells protected from fatty acid-induced cell death. CONCLUSIONS Our results suggest that EC-Atg7 deletion ameliorates HFD-induced obesity and its related metabolic dysfunction, such as insulin resistance and fatty liver by attenuating appetite and vascular rarefaction. The EC-Atg7 deletion may protect the endothelial cells from lipotoxicity and impaired angiogenesis, which preserves the endothelial function in metabolic tissues. These findings may have implications for developing new therapeutic strategies for preventing and treating obesity and its associated health risks.
Collapse
Affiliation(s)
- Guang Ren
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeong-A Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, USA; UAB Comprehensive Diabetes Center, USA.
| |
Collapse
|
5
|
Craig-Schapiro R, Li G, Chen K, Gomez-Salinero JM, Nachman R, Kopacz A, Schreiner R, Chen X, Zhou Q, Rafii S, Redmond D. Single-cell atlas of human pancreatic islet and acinar endothelial cells in health and diabetes. Nat Commun 2025; 16:1338. [PMID: 39915484 PMCID: PMC11802906 DOI: 10.1038/s41467-024-55415-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 12/10/2024] [Indexed: 02/09/2025] Open
Abstract
Characterization of the vascular heterogeneity within the pancreas has previously been lacking. Here, we develop strategies to enrich islet-specific endothelial cells (ISECs) and acinar-specific endothelial cells (ASECs) from three human pancreases and corroborate these findings with three published pancreatic datasets. Single-cell RNA sequencing reveals the unique molecular signatures of ISECs, including structural genes COL13A1, ESM1, PLVAP, UNC5B, and LAMA4, angiocrine genes KDR, THBS1, BMPs and CXCR4, and metabolic genes ACE, PASK and F2RL3. ASECs display distinct signatures including GPIHBP1, CCL14, CD74, AQP1, KLF4, and KLF2, which may manage the inflammatory and metabolic needs of the exocrine pancreas. Ligand-receptor analysis suggests ISECs and ASECs interact with LUM+ fibroblasts and RGS5+ pericytes and smooth muscle cells via VEGF-A:VEGFR2, CXCL12:CXCR4, and LIF:LIFR pathways. Comparative expression and immunohistochemistry indicate disruption of endothelial-expressed CD74, ESM1, PLVAP, THBD, VWA1, and VEGF-A cross-talk among vascular and other cell types in diabetes. Thus, our data provide a single-cell vascular atlas of human pancreas, enabling deeper understanding of pancreatic pathophysiology in health and disease.
Collapse
Affiliation(s)
| | - Ge Li
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Chen
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesus M Gomez-Salinero
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ryan Nachman
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Aleksandra Kopacz
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ryan Schreiner
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xiaojuan Chen
- Columbia Center for Translational Immunology, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Qiao Zhou
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - David Redmond
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Brishti A, Johnson SJ, Palmer DG, Raihan MO, Yan L, Casperson SL. Effects of defined voluntary running distances coupled with high-fat diet consumption on the skeletal muscle transcriptome of male mice. Physiol Rep 2025; 13:e70170. [PMID: 39821584 PMCID: PMC11738645 DOI: 10.14814/phy2.70170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
Exercise counters many adverse health effects of consuming a high-fat diet (HFD). However, complex molecular changes that occur in skeletal muscle in response to exercising while consuming a HFD are not yet known. We investigated the interplay between diverse exercise regimes and HFD consumption on the adaptation of skeletal muscle transcriptome. C57BL/6 male mice were randomized into five groups-one sedentary control group and four exercise groups. The exercise groups consisted of an unrestricted running group (8.3 km/day) and three groups that were restricted to 75%, 50%, or 25% of unrestricted running (6.3, 4.2, and 2.1 km/day, respectively). Total RNA was extracted from frozen gastrocnemius muscle for transcriptome analyses. DEG counts were 1347, 1823, 1103, and 1107 and there were 107, 169, 67, and 89 unique genes present in the HFD-25%, HFD-50%, HFD-75%, and HFD-U, respectively. Comparing exercise groups, we found that exercising at 50% resulted in the most differentially expressed transcripts with the MAPK and PPAR signaling pathways enriched in down- and up-regulated genes, respectively. These results demonstrate that running distance impacts the adaptation of the skeletal muscle transcriptome to exercise and suggest that middle-distance running may provide the greatest protection against high-fat diet-induced stress coupled with exercise.
Collapse
Affiliation(s)
- Afrina Brishti
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Sarah J. Johnson
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
- Present address:
Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Daniel G. Palmer
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Md Obayed Raihan
- Department of Pharmaceutical Sciences, College of Health Sciences and PharmacyChicago State UniversityChicagoIllinoisUSA
| | - Lin Yan
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| | - Shanon L. Casperson
- United States Department of Agriculture, Agricultural Research ServiceGrand Forks Human Nutrition Research CenterGrand ForksNorth DakotaUSA
| |
Collapse
|
7
|
Lu X, Xie Q, Pan X, Zhang R, Zhang X, Peng G, Zhang Y, Shen S, Tong N. Type 2 diabetes mellitus in adults: pathogenesis, prevention and therapy. Signal Transduct Target Ther 2024; 9:262. [PMID: 39353925 PMCID: PMC11445387 DOI: 10.1038/s41392-024-01951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Type 2 diabetes (T2D) is a disease characterized by heterogeneously progressive loss of islet β cell insulin secretion usually occurring after the presence of insulin resistance (IR) and it is one component of metabolic syndrome (MS), and we named it metabolic dysfunction syndrome (MDS). The pathogenesis of T2D is not fully understood, with IR and β cell dysfunction playing central roles in its pathophysiology. Dyslipidemia, hyperglycemia, along with other metabolic disorders, results in IR and/or islet β cell dysfunction via some shared pathways, such as inflammation, endoplasmic reticulum stress (ERS), oxidative stress, and ectopic lipid deposition. There is currently no cure for T2D, but it can be prevented or in remission by lifestyle intervention and/or some medication. If prevention fails, holistic and personalized management should be taken as soon as possible through timely detection and diagnosis, considering target organ protection, comorbidities, treatment goals, and other factors in reality. T2D is often accompanied by other components of MDS, such as preobesity/obesity, metabolic dysfunction associated steatotic liver disease, dyslipidemia, which usually occurs before it, and they are considered as the upstream diseases of T2D. It is more appropriate to call "diabetic complications" as "MDS-related target organ damage (TOD)", since their development involves not only hyperglycemia but also other metabolic disorders of MDS, promoting an up-to-date management philosophy. In this review, we aim to summarize the underlying mechanism, screening, diagnosis, prevention, and treatment of T2D, especially regarding the personalized selection of hypoglycemic agents and holistic management based on the concept of "MDS-related TOD".
Collapse
Affiliation(s)
- Xi Lu
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxing Xie
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohui Pan
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Ruining Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Ge Peng
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Yuwei Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Sumin Shen
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Taylor J, Uhl L, Moll I, Hasan SS, Wiedmann L, Morgenstern J, Giaimo BD, Friedrich T, Alsina-Sanchis E, De Angelis Rigotti F, Mülfarth R, Kaltenbach S, Schenk D, Nickel F, Fleming T, Sprinzak D, Mogler C, Korff T, Billeter AT, Müller-Stich BP, Berriel Diaz M, Borggrefe T, Herzig S, Rohm M, Rodriguez-Vita J, Fischer A. Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia. NATURE CANCER 2023; 4:1544-1560. [PMID: 37749321 PMCID: PMC10663158 DOI: 10.1038/s43018-023-00622-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/20/2023] [Indexed: 09/27/2023]
Abstract
Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals.
Collapse
Affiliation(s)
- Jacqueline Taylor
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leonie Uhl
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sana Safatul Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Friedrich
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Giessen, Germany
| | - Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Francesca De Angelis Rigotti
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Kaltenbach
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Darius Schenk
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Nickel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adrian T Billeter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
9
|
Hasan SS, Fischer A. Notch Signaling in the Vasculature: Angiogenesis and Angiocrine Functions. Cold Spring Harb Perspect Med 2023; 13:a041166. [PMID: 35667708 PMCID: PMC9899647 DOI: 10.1101/cshperspect.a041166] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Formation of a functional blood vessel network is a complex process tightly controlled by pro- and antiangiogenic signals released within the local microenvironment or delivered through the bloodstream. Endothelial cells precisely integrate such temporal and spatial changes in extracellular signals and generate an orchestrated response by modulating signaling transduction, gene expression, and metabolism. A key regulator in vessel formation is Notch signaling, which controls endothelial cell specification, proliferation, migration, adhesion, and arteriovenous differentiation. This review summarizes the molecular biology of endothelial Notch signaling and how it controls angiogenesis and maintenance of the established, quiescent vasculature. In addition, recent progress in the understanding of Notch signaling in endothelial cells for controlling organ homeostasis by transcriptional regulation of angiocrine factors and its relevance to disease will be discussed.
Collapse
Affiliation(s)
- Sana S Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
10
|
Zhou W, Yu G, Liu L, Gao Q, Feng L, Wang Y. Primary diffuse large B-cell lymphoma of the breast: A retrospective study of outcomes and insulin resistance. Saudi Med J 2023; 44:38-44. [PMID: 36634942 PMCID: PMC9987672 DOI: 10.15537/smj.2023.44.1.20220677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/14/2022] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES To investigate the clinicopathological features, insulin resistance (IR) status, and the outcomes of populations with diffuse large B-cell lymphoma (DLBCL) of the breast. METHODS This study was carried out at Department of Haematology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China, that included 32 patients treated form January 2009 to June 2020. The primary endpoints of the study were their survival time. RESULTS There were 32 patients in the study. A total of 18 (56.2%) patients had IR. In terms of treatment, 31.2% were treated with surgery, most (93.8%) received chemotherapy, and 25% received radiotherapy and intrathecal therapy. Univariate analysis indicated the patients with stages III-IV, B symptoms, tumour recurrence, PAX5 positivity, and c-MYC positivity showed a shorter survival time (p<0.05). The overall survival and progression-free survival (PFS) rates in IR group were shorter than those without IR, but there was no statistical difference (p>0.05). Multivariate analysis indicated that tumour recurrence shortened the 5-year PFS of the patients (p=0.037). CONCLUSION Primary DLBCL of the breast was very rare; more than half of the cases had IR, but IR did not affect their survival.
Collapse
Affiliation(s)
- Weiling Zhou
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
| | - Guodong Yu
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
| | - Lihong Liu
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
| | - Qian Gao
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
| | - Lei Feng
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
| | - Yuan Wang
- From the Department of Endocrine and Metabolic Diseases (Zhou, Gao, Feng, Wang), from the Department of Hematology (Liu), The Fourth Hospital of Hebei Medical University, Shijiazhuang, and from the Department of Hepatobiliary Surgery (Yu), Affiliated Hospital of Hebei University, Baoding, China.
- Address correspondence and reprint request to: Dr. Yuan Wang, Department of Endocrine and Metabolic Diseases, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China. E-mail: ORCID ID: https://orcid.org/0000-0003-1657-1484
| |
Collapse
|
11
|
Alsina-Sanchis E, Mülfarth R, Moll I, Böhn S, Wiedmann L, Jordana-Urriza L, Ziegelbauer T, Zimmer E, Taylor J, De Angelis Rigotti F, Stögbauer A, Giaimo BD, Cerwenka A, Borggrefe T, Fischer A, Rodriguez-Vita J. Endothelial RBPJ Is Essential for the Education of Tumor-Associated Macrophages. Cancer Res 2022; 82:4414-4428. [PMID: 36200806 DOI: 10.1158/0008-5472.can-22-0076] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 08/03/2022] [Accepted: 09/30/2022] [Indexed: 01/24/2023]
Abstract
Epithelial ovarian cancer (EOC) is one of the most lethal gynecologic cancers worldwide. EOC cells educate tumor-associated macrophages (TAM) through CD44-mediated cholesterol depletion to generate an immunosuppressive tumor microenvironment (TME). In addition, tumor cells frequently activate Notch1 receptors on endothelial cells (EC) to facilitate metastasis. However, further work is required to establish whether the endothelium also influences the education of recruited monocytes. Here, we report that canonical Notch signaling through RBPJ in ECs is an important player in the education of TAMs and EOC progression. Deletion of Rbpj in the endothelium of adult mice reduced infiltration of monocyte-derived macrophages into the TME of EOC and prevented the acquisition of a typical TAM gene signature; this was associated with stronger cytotoxic activity of T cells and decreased tumor burden. Mechanistically, CXCL2 was identified as a novel Notch/RBPJ target gene that regulated the expression of CD44 on monocytes and subsequent cholesterol depletion of TAMs. Bioinformatic analysis of ovarian cancer patient data showed that increased CXCL2 expression is accompanied by higher expression of CD44 and TAM education. Together, these findings indicate that EOC cells induce the tumor endothelium to secrete CXCL2 to establish an immunosuppressive microenvironment. SIGNIFICANCE Endothelial Notch signaling favors immunosuppression by increasing CXCL2 secretion to stimulate CD44 expression in macrophages, facilitating their education by tumor cells.
Collapse
Affiliation(s)
- Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Böhn
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Lorea Jordana-Urriza
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tara Ziegelbauer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eleni Zimmer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jacqueline Taylor
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesca De Angelis Rigotti
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumour-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Adrian Stögbauer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany Tissue
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumour-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
12
|
Abstract
Notch signalling is an evolutionarily highly conserved signalling mechanism governing differentiation and regulating homeostasis in many tissues. In this review, we discuss recent advances in our understanding of the roles that Notch signalling plays in the vasculature. We describe how Notch signalling regulates different steps during the genesis and remodelling of blood vessels (vasculogenesis and angiogenesis), including critical roles in assigning arterial and venous identities to the emerging blood vessels and regulation of their branching. We then proceed to discuss how experimental perturbation of Notch signalling in the vasculature later in development affects vascular homeostasis. In this review, we also describe how dysregulated Notch signalling, as a consequence of direct mutations of genes in the Notch pathway or aberrant Notch signalling output, contributes to various types of vascular disease, including CADASIL, Snedden syndrome and pulmonary arterial hypertension. Finally, we point out some of the current knowledge gaps and identify remaining challenges in understanding the role of Notch in the vasculature, which need to be addressed to pave the way for Notch-based therapies to cure or ameliorate vascular disease.
Collapse
Affiliation(s)
- Francesca Del Gaudio
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Dongli Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden,Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
13
|
O’Brien KA, Murray AJ, Simonson TS. Notch Signaling and Cross-Talk in Hypoxia: A Candidate Pathway for High-Altitude Adaptation. Life (Basel) 2022; 12:437. [PMID: 35330188 PMCID: PMC8954738 DOI: 10.3390/life12030437] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 12/17/2022] Open
Abstract
Hypoxia triggers complex inter- and intracellular signals that regulate tissue oxygen (O2) homeostasis, adjusting convective O2 delivery and utilization (i.e., metabolism). Human populations have been exposed to high-altitude hypoxia for thousands of years and, in doing so, have undergone natural selection of multiple gene regions supporting adaptive traits. Some of the strongest selection signals identified in highland populations emanate from hypoxia-inducible factor (HIF) pathway genes. The HIF pathway is a master regulator of the cellular hypoxic response, but it is not the only regulatory pathway under positive selection. For instance, regions linked to the highly conserved Notch signaling pathway are also top targets, and this pathway is likely to play essential roles that confer hypoxia tolerance. Here, we explored the importance of the Notch pathway in mediating the cellular hypoxic response. We assessed transcriptional regulation of the Notch pathway, including close cross-talk with HIF signaling, and its involvement in the mediation of angiogenesis, cellular metabolism, inflammation, and oxidative stress, relating these functions to generational hypoxia adaptation.
Collapse
Affiliation(s)
- Katie A. O’Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK;
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK;
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
14
|
Abstract
Obesity has reached epidemic proportions and is a major contributor to insulin resistance (IR) and type 2 diabetes (T2D). Importantly, IR and T2D substantially increase the risk of cardiovascular (CV) disease. Although there are successful approaches to maintain glycemic control, there continue to be increased CV morbidity and mortality associated with metabolic disease. Therefore, there is an urgent need to understand the cellular and molecular processes that underlie cardiometabolic changes that occur during obesity so that optimal medical therapies can be designed to attenuate or prevent the sequelae of this disease. The vascular endothelium is in constant contact with the circulating milieu; thus, it is not surprising that obesity-driven elevations in lipids, glucose, and proinflammatory mediators induce endothelial dysfunction, vascular inflammation, and vascular remodeling in all segments of the vasculature. As cardiometabolic disease progresses, so do pathological changes in the entire vascular network, which can feed forward to exacerbate disease progression. Recent cellular and molecular data have implicated the vasculature as an initiating and instigating factor in the development of several cardiometabolic diseases. This Review discusses these findings in the context of atherosclerosis, IR and T2D, and heart failure with preserved ejection fraction. In addition, novel strategies to therapeutically target the vasculature to lessen cardiometabolic disease burden are introduced.
Collapse
|
15
|
Developmental angiocrine diversification of endothelial cells for organotypic regeneration. Dev Cell 2021; 56:3042-3051. [PMID: 34813766 DOI: 10.1016/j.devcel.2021.10.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.
Collapse
|
16
|
Abstract
The endothelium acts as the barrier that prevents circulating lipids such as lipoproteins and fatty acids into the arterial wall; it also regulates normal functioning in the circulatory system by balancing vasodilation and vasoconstriction, modulating the several responses and signals. Plasma lipids can interact with endothelium via different mechanisms and produce different phenotypes. Increased plasma-free fatty acids (FFAs) levels are associated with the pathogenesis of atherosclerosis and cardiovascular diseases (CVD). Because of the multi-dimensional roles of plasma FFAs in mediating endothelial dysfunction, increased FFA level is now considered an essential link in the onset of endothelial dysfunction in CVD. FFA-mediated endothelial dysfunction involves several mechanisms, including dysregulated production of nitric oxide and cytokines, metaflammation, oxidative stress, inflammation, activation of the renin-angiotensin system, and apoptosis. Therefore, modulation of FFA-mediated pathways involved in endothelial dysfunction may prevent the complications associated with CVD risk. This review presents details as to how endothelium is affected by FFAs involving several metabolic pathways.
Collapse
|
17
|
Fatty acids and evolving roles of their proteins in neurological, cardiovascular disorders and cancers. Prog Lipid Res 2021; 83:101116. [PMID: 34293403 DOI: 10.1016/j.plipres.2021.101116] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/04/2021] [Accepted: 07/14/2021] [Indexed: 01/03/2023]
Abstract
The dysregulation of fat metabolism is involved in various disorders, including neurodegenerative, cardiovascular, and cancers. The uptake of long-chain fatty acids (LCFAs) with 14 or more carbons plays a pivotal role in cellular metabolic homeostasis. Therefore, the uptake and metabolism of LCFAs must constantly be in tune with the cellular, metabolic, and structural requirements of cells. Many metabolic diseases are thought to be driven by the abnormal flow of fatty acids either from the dietary origin and/or released from adipose stores. Cellular uptake and intracellular trafficking of fatty acids are facilitated ubiquitously with unique combinations of fatty acid transport proteins and cytoplasmic fatty acid-binding proteins in every tissue. Extensive data are emerging on the defective transporters and metabolism of LCFAs and their clinical implications. Uptake and metabolism of LCFAs are crucial for the brain's functional development and cardiovascular health and maintenance. In addition, data suggest fatty acid metabolic transporter can normalize activated inflammatory response by reprogramming lipid metabolism in cancers. Here we review the current understanding of how LCFAs and their proteins contribute to the pathophysiology of three crucial diseases and the mechanisms involved in the processes.
Collapse
|
18
|
Fu J, Yu MG, Li Q, Park K, King GL. Insulin's actions on vascular tissues: Physiological effects and pathophysiological contributions to vascular complications of diabetes. Mol Metab 2021; 52:101236. [PMID: 33878400 PMCID: PMC8513152 DOI: 10.1016/j.molmet.2021.101236] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background Insulin has been demonstrated to exert direct and indirect effects on vascular tissues. Its actions in vascular cells are mediated by two major pathways: the insulin receptor substrate 1/2-phosphoinositide-3 kinase/Akt (IRS1/2/PI3K/Akt) pathway and the Src/mitogen-activated protein kinase (MAPK) pathway, both of which contribute to the expression and distribution of metabolites, hormones, and cytokines. Scope of review In this review, we summarize the current understanding of insulin's physiological and pathophysiological actions and associated signaling pathways in vascular cells, mainly in endothelial cells (EC) and vascular smooth muscle cells (VSMC), and how these processes lead to selective insulin resistance. We also describe insulin's potential new signaling and biological effects derived from animal studies and cultured capillary and arterial EC, VSMC, and pericytes. We will not provide a detailed discussion of insulin's effects on the myocardium, insulin's structure, or its signaling pathways' various steps, since other articles in this issue discuss these areas in depth. Major conclusions Insulin mediates many important functions on vascular cells via its receptors and signaling cascades. Its direct actions on EC and VSMC are important for transporting and communicating nutrients, cytokines, hormones, and other signaling molecules. These vascular actions are also important for regulating systemic fuel metabolism and energetics. Inhibiting or enhancing these pathways leads to selective insulin resistance, exacerbating the development of endothelial dysfunction, atherosclerosis, restenosis, poor wound healing, and even myocardial dysfunction. Targeted therapies to improve selective insulin resistance in EC and VSMC are thus needed to specifically mitigate these pathological processes. Insulin's actions in vascular cells have a significant influence on systemic metabolism. Insulin exerts its vascular effects through its receptors and signaling cascades. Inhibition or enhancement of different insulin signaling leads to selective insulin resistance. Loss of insulin's actions causes endothelial dysfunction and vascular complications in diabetes.
Collapse
Affiliation(s)
- Jialin Fu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Marc Gregory Yu
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Qian Li
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - George L King
- Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Trans-endothelial trafficking of metabolic substrates and its importance in cardio-metabolic disease. Biochem Soc Trans 2021; 49:507-517. [PMID: 33616631 DOI: 10.1042/bst20200991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 11/17/2022]
Abstract
The endothelium acts as a gatekeeper, controlling the movement of biomolecules between the circulation and underlying tissues. Although conditions of metabolic stress are traditionally considered as causes of endothelial dysfunction, a principal driver of cardiovascular disease, accumulating evidence suggests that endothelial cells are also active players in maintaining local metabolic homeostasis, in part, through regulating the supply of metabolic substrates, including lipids and glucose, to energy-demanding organs. Therefore, endothelial dysfunction, in terms of altered trans-endothelial trafficking of these substrates, may in fact be an early contributor towards the establishment of metabolic dysfunction and subsequent cardiovascular disease. Understanding the molecular mechanisms that underpin substrate trafficking through the endothelium represents an important area within the vascular and metabolism fields that may offer an opportunity for identifying novel therapeutic targets. This mini-review summarises the emerging mechanisms regulating the trafficking of lipids and glucose through the endothelial barrier and how this may impact on the development of cardio-metabolic disease.
Collapse
|
20
|
Herold J, Kalucka J. Angiogenesis in Adipose Tissue: The Interplay Between Adipose and Endothelial Cells. Front Physiol 2021; 11:624903. [PMID: 33633579 PMCID: PMC7900516 DOI: 10.3389/fphys.2020.624903] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is a worldwide health problem, and as its prevalence increases, so does the burden of obesity-associated co-morbidities like type 2 diabetes or cardiovascular diseases (CVDs). Adipose tissue (AT) is an endocrine organ embedded in a dense vascular network. AT regulates the production of hormones, angiogenic factors, and cytokines. During the development of obesity, AT expands through the increase in fat cell size (hypertrophy) and/or fat cell number (hyperplasia). The plasticity and expansion of AT is related to its angiogenic capacities. Angiogenesis is a tightly orchestrated process, which involves endothelial cell (EC) proliferation, migration, invasion, and new tube formation. The expansion of AT is accelerated by hypoxia, inflammation, and structural remodeling of blood vessels. The paracrine signaling regulates the functional link between ECs and adipocytes. Adipocytes can secrete both pro-angiogenic molecules, e.g., tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), or vascular endothelial growth factor (VEGF), and anti-angiogenic factors, e.g., serpins. If the pro-angiogenic molecules dominate, the angiogenesis is dysregulated and the endothelium becomes dysfunctional. However, if anti-angiogenic molecules are overexpressed relative to the angiogenic regulators, the angiogenesis is repressed, and AT becomes hypoxic. Furthermore, in the presence of chronic nutritional excess, endothelium loses its primary function and contributes to the inflammation and fibrosis of AT, which increases the risk for CVDs. This review discusses the current understanding of ECs function in AT, the cross-talk between adipose and ECs, and how obesity can lead to its dysfunction. Understanding the interplay of angiogenesis with AT can be an approach to therapy obesity and obesity-related diseases such as CVDs.
Collapse
Affiliation(s)
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Hasan SS, Fischer A. The Endothelium: An Active Regulator of Lipid and Glucose Homeostasis. Trends Cell Biol 2020; 31:37-49. [PMID: 33129632 DOI: 10.1016/j.tcb.2020.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
The vascular endothelium serves as a dynamic barrier that separates blood from interstitia. Endothelial cells (ECs) respond rapidly to changes in the circulation and actively regulate vessel tone, permeability, and platelet functions. ECs also secrete angiocrine factors that dictate the function of adjacent parenchymal cells in an organ-specific manner. Endothelial dysfunction is considered as a hallmark of metabolic diseases. However, there is emerging evidence that ECs modulate the transfer of nutrients and hormones to parenchymal cells in response to alterations in metabolic profile. As such, a causal role for ECs in systemic metabolic dysregulation can be envisaged. This review summarizes recent progress in the understanding of regulated fatty acid, glucose, and insulin transport across the endothelium and discusses its pathophysiological implications.
Collapse
Affiliation(s)
- Sana S Hasan
- Division of Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Fischer
- Division of Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, 69120 Heidelberg, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
22
|
Filippini A, D’Alessio A. Caveolae and Lipid Rafts in Endothelium: Valuable Organelles for Multiple Functions. Biomolecules 2020; 10:biom10091218. [PMID: 32825713 PMCID: PMC7563503 DOI: 10.3390/biom10091218] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022] Open
Abstract
Caveolae are flask-shaped invaginations of the plasma membrane found in numerous cell types and are particularly abundant in endothelial cells and adipocytes. The lipid composition of caveolae largely matches that of lipid rafts microdomains that are particularly enriched in cholesterol, sphingomyelin, glycosphingolipids, and saturated fatty acids. Unlike lipid rafts, whose existence remains quite elusive in living cells, caveolae can be clearly distinguished by electron microscope. Despite their similar composition and the sharing of some functions, lipid rafts appear more heterogeneous in terms of size and are more dynamic than caveolae. Following the discovery of caveolin-1, the first molecular marker as well as the unique scaffolding protein of caveolae, we have witnessed a remarkable increase in studies aimed at investigating the role of these organelles in cell functions and human disease. The goal of this review is to discuss the most recent studies related to the role of caveolae and caveolins in endothelial cells. We first recapitulate the major embryological processes leading to the formation of the vascular tree. We next discuss the contribution of caveolins and cavins to membrane biogenesis and cell response to extracellular stimuli. We also address how caveolae and caveolins control endothelial cell metabolism, a central mechanism involved in migration proliferation and angiogenesis. Finally, as regards the emergency caused by COVID-19, we propose to study the caveolar platform as a potential target to block virus entry into endothelial cells.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Roma, Italy;
| | - Alessio D’Alessio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italia
- Correspondence:
| |
Collapse
|
23
|
Hasan SS, Jabs M, Taylor J, Wiedmann L, Leibing T, Nordström V, Federico G, Roma LP, Carlein C, Wolff G, Ekim-Üstünel B, Brune M, Moll I, Tetzlaff F, Gröne HJ, Fleming T, Géraud C, Herzig S, Nawroth PP, Fischer A. Endothelial Notch signaling controls insulin transport in muscle. EMBO Mol Med 2020; 12:e09271. [PMID: 32187826 PMCID: PMC7136962 DOI: 10.15252/emmm.201809271] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
The role of the endothelium is not just limited to acting as an inert barrier for facilitating blood transport. Endothelial cells (ECs), through expression of a repertoire of angiocrine molecules, regulate metabolic demands in an organ‐specific manner. Insulin flux across the endothelium to muscle cells is a rate‐limiting process influencing insulin‐mediated lowering of blood glucose. Here, we demonstrate that Notch signaling in ECs regulates insulin transport to muscle. Notch signaling activity was higher in ECs isolated from obese mice compared to non‐obese. Sustained Notch signaling in ECs lowered insulin sensitivity and increased blood glucose levels. On the contrary, EC‐specific inhibition of Notch signaling increased insulin sensitivity and improved glucose tolerance and glucose uptake in muscle in a high‐fat diet‐induced insulin resistance model. This was associated with increased transcription of Cav1, Cav2, and Cavin1, higher number of caveolae in ECs, and insulin uptake rates, as well as increased microvessel density. These data imply that Notch signaling in the endothelium actively controls insulin sensitivity and glucose homeostasis and may therefore represent a therapeutic target for diabetes.
Collapse
Affiliation(s)
- Sana S Hasan
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Jabs
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jacqueline Taylor
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Thomas Leibing
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Section of Clinical and Molecular Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Viola Nordström
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Giuseppina Federico
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Christopher Carlein
- Biophysics Department, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Gretchen Wolff
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Bilgen Ekim-Üstünel
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Maik Brune
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Tetzlaff
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann-Josef Gröne
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pharmacology, Philipps University of Marburg, Marburg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Section of Clinical and Molecular Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Peter P Nawroth
- Institute for Diabetes and Cancer (IDC) and Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|