1
|
Iwase T, Parikh A, Wenli D, Shen Y, Adams DL, Tang CM, Cohen EN, Reuben JM, Shrimanker TV, Chainitikun S, Kida K, Raghavendra AS, Sapon ME, Sahin O, James A, Sridhar N, Klopp AH, Tripathy D, Ueno NT. Circulating cancer-associated macrophage-like cells and macrophage-related cytokines in obese patients with advanced breast cancer who undergo neoadjuvant chemotherapy. J Cancer 2024; 15:5855-5862. [PMID: 39440056 PMCID: PMC11493001 DOI: 10.7150/jca.89453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/05/2024] [Indexed: 10/25/2024] Open
Abstract
Purpose: Cancer-associated macrophage-like cells (CAMLs) are rare, gigantic, and atypical circulating cells found exclusively in the peripheral blood of patients with solid cancers. Obesity-induced hypoxia attracts macrophages to the tumor microenvironment, where they contribute to establishing chronic inflammation, leading to cancer progression. We hypothesized that obese patients with advanced breast cancer may have CAML profiles different from those of nonobese patients, and these profiles may correlate with proinflammatory markers or other macrophage-related markers. Methods: We prospectively collected 20 mL of peripheral blood from patients diagnosed with stage 2-4 breast cancer. We identified CAMLs using the CellSieve microfiltration system and in parallel quantified the proinflammatory and macrophage-related markers using a multiplex cytokine panel. We further evaluated C-X-C chemokine receptor type 4 (CXCR4) expression in CAMLs to investigate its relationship to the macrophage differentiation. We estimated the association between CAML characteristics and body mass index (BMI), body composition, and cytokines/chemokines. Results: Thirty patients were included in the study, and 28 samples were analyzed. Higher BMI was significantly correlated with the increased maximum CAML size (P = 0.035). Patients with higher BMIs had significantly increased macrophage-colony stimulating factor (M-CSF) levels in plasma (P = 0.007), and obese patients trended towards higher tumor necrosis factor-alpha, MIP-1α and M-CSF expression (P <0.10). Body composition analysis showed that the M-CSF and SAT amounts were significantly correlated (P = 0.010). MIP-1α expression was significantly correlated with average CXCR4 CAML expression (P = 0.003). Conclusion: We discovered larger CAML size was associated with SAT-dominant obesity with increased macrophage-related and proinflammatory markers in obese than in nonobese breast cancer patients.
Collapse
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- University of Hawai'i Cancer Center, 701 Ilalo Street Honolulu, HI 96813, USA
| | - Aaroh Parikh
- Diagnostic Radiology, Baylor College of Medicine, 1 Moursund St, Houston, TX 77030, USA
| | - Dong Wenli
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Daniel L. Adams
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Cha-mei Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Evan N. Cohen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | - Sudpreeda Chainitikun
- Department of Medical Oncology, MedPark Hospital, 3333 Rama IV Rd, Khlong Toei, Bangkok 10110, Thailand
| | - Kumiko Kida
- Breast Center, St. Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
| | - Akshara Singareeka Raghavendra
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Maryanne E. Sapon
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Onur Sahin
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Anjali James
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nithya Sridhar
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ann H. Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Debasish Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Naoto T. Ueno
- University of Hawai'i Cancer Center, 701 Ilalo Street Honolulu, HI 96813, USA
| |
Collapse
|
2
|
Wang Q, Hartig SM, Ballantyne CM, Wu H. The multifaceted life of macrophages in white adipose tissue: Immune shift couples with metabolic switch. Immunol Rev 2024; 324:11-24. [PMID: 38683173 PMCID: PMC11262992 DOI: 10.1111/imr.13338] [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] [Indexed: 05/01/2024]
Abstract
White adipose tissue (WAT) is a vital endocrine organ that regulates energy balance and metabolic homeostasis. In addition to fat cells, WAT harbors macrophages with distinct phenotypes that play crucial roles in immunity and metabolism. Nutrient demands cause macrophages to accumulate in WAT niches, where they remodel the microenvironment and produce beneficial or detrimental effects on systemic metabolism. Given the abundance of macrophages in WAT, this review summarizes the heterogeneity of WAT macrophages in physiological and pathological conditions, including their alterations in quantity, phenotypes, characteristics, and functions during WAT growth and development, as well as healthy or unhealthy expansion. We will discuss the interactions of macrophages with other cell partners in WAT including adipose stem cells, adipocytes, and T cells in the context of various microenvironment niches in lean or obese condition. Finally, we highlight how adipose tissue macrophages merge immunity and metabolic changes to govern energy balance for the organism.
Collapse
Affiliation(s)
- Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Sean M. Hartig
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA 77030
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA 77030
| |
Collapse
|
3
|
van den Munckhof ICL, Bahrar H, Schraa K, Brand T, Ter Horst R, van der Graaf M, Dekker HM, Stienstra R, de Graaf J, Joosten LAB, Netea MG, Riksen NP, Rutten JHW. Sex-specific association of visceral and subcutaneous adipose tissue volumes with systemic inflammation and innate immune cells in people living with obesity. Int J Obes (Lond) 2024; 48:523-532. [PMID: 38135702 DOI: 10.1038/s41366-023-01444-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND AIMS Obesity predisposes to metabolic and cardiovascular diseases. Adipose tissue inflammation and systemic inflammation contribute to these complications. There are strong sex differences in adipose tissue distribution and in systemic inflammation. Women have more subcutaneous adipose tissue (SAT) and less visceral adipose tissue (VAT) than men. We explored the sex differences in the association between the different adipose compartments and inflammatory markers that are important in cardiometabolic disease pathophysiology. METHODS Single-center observational cohort study with 302 individuals with a BMI ≥ 27 kg/m2. We were unable to acquire MRI data from seven individuals and from another 18 the MRI data were not usable, resulting in 277 people (155 men, 122 women), aged 55-81 years. INTERVENTION We performed the following measurements: abdominal magnetic resonance imaging to measure VAT, and SAT (deep and superficial) volumes; circulating leukocyte counts and cytokine production capacity of peripheral blood mononuclear cells (PBMCs), circulating cytokines, adipokines, and targeted proteomics; abdominal sSAT biopsies for histology and gene expression. RESULTS Only in women, (s)SAT volume was associated with circulating leukocytes, monocytes, and neutrophils. Circulating IL-6 and IL-18BP were associated with SAT volume in women and VAT in men. Several circulating proteins, including monocyte-colony-stimulating factor 1 and hepatocyte growth factor, are associated with sSAT in women and VAT in men. Only in women, SAT volume is associated with SAT expression of inflammatory proteins, including leptin, CD68, TNFα and IL-1α. CONCLUSION In women living with obesity, abdominal SAT volume, especially sSAT, is associated with circulating leukocytes and inflammatory proteins. In men, these parameters mainly show associations with VAT volume. This could be because only in women, sSAT volume is associated with sSAT expression of inflammatory proteins. These findings underscore that future research on adipose tissue in relation to cardiometabolic and cardiovascular disease should take sex differences into account.
Collapse
Affiliation(s)
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kiki Schraa
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tessa Brand
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob Ter Horst
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Helena M Dekker
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Jacqueline de Graaf
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
4
|
Sarsani V, Brotman SM, Xianyong Y, Fernandes Silva L, Laakso M, Spracklen CN. A cross-ancestry genome-wide meta-analysis, fine-mapping, and gene prioritization approach to characterize the genetic architecture of adiponectin. HGG ADVANCES 2024; 5:100252. [PMID: 37859345 PMCID: PMC10652123 DOI: 10.1016/j.xhgg.2023.100252] [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: 06/29/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023] Open
Abstract
Previous genome-wide association studies (GWASs) for adiponectin, a complex trait linked to type 2 diabetes and obesity, identified >20 associated loci. However, most loci were identified in populations of European ancestry, and many of the target genes underlying the associations remain unknown. We conducted a cross-ancestry adiponectin GWAS meta-analysis in ≤46,434 individuals from the Metabolic Syndrome in Men (METSIM) cohort and the ADIPOGen and AGEN consortiums. We combined study-specific association summary statistics using a fixed-effects, inverse variance-weighted approach. We identified 22 loci associated with adiponectin (p < 5×10-8), including 15 known and seven previously unreported loci. Among individuals of European ancestry, Genome-wide Complex Traits Analysis joint conditional analysis (GCTA-COJO) identified 14 additional distinct signals at the ADIPOQ, CDH13, HCAR1, and ZNF664 loci. Leveraging the cross-ancestry data, FINEMAP + SuSiE identified 45 causal variants (PP > 0.9), which also exhibited potential pleiotropy for cardiometabolic traits. To prioritize target genes at associated loci, we propose a combinatorial likelihood scoring formalism (Gene Priority Score [GPScore]) based on measures derived from 11 gene prioritization strategies and the physical distance to the transcription start site. With GPScore, we prioritize the 30 most probable target genes underlying the adiponectin-associated variants in the cross-ancestry analysis, including well-known causal genes (e.g., ADIPOQ, CDH13) and additional genes (e.g., CSF1, RGS17). Functional association networks revealed complex interactions of prioritized genes, their functionally connected genes, and their underlying pathways centered around insulin and adiponectin signaling, indicating an essential role in regulating energy balance in the body, inflammation, coagulation, fibrinolysis, insulin resistance, and diabetes. Overall, our analyses identify and characterize adiponectin association signals and inform experimental interrogation of target genes for adiponectin.
Collapse
Affiliation(s)
- Vishal Sarsani
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, USA
| | - Sarah M Brotman
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yin Xianyong
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Lillian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
5
|
Park JM, Kim J, Lee YJ, Bae SU, Lee HW. Inflammatory bowel disease-associated intestinal fibrosis. J Pathol Transl Med 2023; 57:60-66. [PMID: 36623814 PMCID: PMC9846010 DOI: 10.4132/jptm.2022.11.02] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 01/11/2023] Open
Abstract
Fibrosis is characterized by a proliferation of fibroblasts and excessive extracellular matrix following chronic inflammation, and this replacement of organ tissue with fibrotic tissue causes a loss of function. Inflammatory bowel disease (IBD) is a chronic inflammation of the gastrointestinal tract, and intestinal fibrosis is common in IBD patients, resulting in several complications that require surgery, such as a stricture or penetration. This review describes the pathogenesis and various factors involved in intestinal fibrosis in IBD, including cytokines, growth factors, epithelial-mesenchymal and endothelial-mesenchymal transitions, and gut microbiota. Furthermore, histopathologic findings and scoring systems used for stenosis in IBD are discussed, and differences in the fibrosis patterns of ulcerative colitis and Crohn's disease are compared. Biomarkers and therapeutic agents targeting intestinal fibrosis are briefly mentioned at the end.
Collapse
Affiliation(s)
- Ji Min Park
- Department of Pathology, Keimyung University School of Medicine, Daegu,
Korea
| | - Jeongseok Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu,
Korea
| | - Yoo Jin Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu,
Korea
| | - Sung Uk Bae
- Division of Colorectal Surgery, Department of Surgery, Keimyung University School of Medicine, Daegu,
Korea
| | - Hye Won Lee
- Department of Pathology, Keimyung University School of Medicine, Daegu,
Korea
| |
Collapse
|
6
|
Perry AS, Tanriverdi K, Risitano A, Hwang SJ, Murthy VL, Nayor M, Zhao S, Levy D, Shah RV, Freedman JE. The inflammatory proteome, obesity, and medical weight loss and regain in humans. Obesity (Silver Spring) 2023; 31:150-158. [PMID: 36334095 PMCID: PMC9923277 DOI: 10.1002/oby.23587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Weight regain occurs after medical weight loss via mechanisms of post-weight-loss "metabolic adaptation." The relationship of inflammatory proteins with weight loss/regain was studied to determine a role for inflammation in metabolic adaptation. METHODS Seventy-four proteins central to inflammation and immune regulation (Olink) were analyzed in plasma from up to 490 participants in a trial of medical weight-loss maintenance. Cross-sectional and longitudinal associations of proteins with weight were measured using linear and mixed effects regression models and t testing, with replication in the Framingham Heart Study. RESULTS Broad changes in the inflammatory proteome were observed among the study cohort (60% women, 35% African American) with initial weight loss of ≈8 kg from a median 94 kg at study entry (33/74 proteins; 7 increased; 26 decreased), many of which tracked with weight regain of median ≈2 kg over the next 30 months. Ten proteins were associated with different rates of weight regain, some specifying pathways of chemotaxis and innate immune responses. Several of the observed protein associations were also linked to prevalent obesity in the Framingham Heart Study. CONCLUSIONS Broad changes in the inflammatory proteome track with changes in weight and may identify specific pathways that modify patterns of weight regain.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Antonina Risitano
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Shih-Jen Hwang
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Venkatesh L Murthy
- Department of Medicine and Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew Nayor
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Zhai G, Pang Y, Zou Y, Wang X, Liu J, Zhang Q, Cao Z, Wang N, Li H, Wang Y. Effects of PLIN1 Gene Knockout on the Proliferation, Apoptosis, Differentiation and Lipolysis of Chicken Preadipocytes. Animals (Basel) 2022; 13:92. [PMID: 36611701 PMCID: PMC9817814 DOI: 10.3390/ani13010092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Perilipin 1 (PLIN1) is one of the most abundant lipid droplet-related proteins on the surface of adipocytes. Our previous results showed that PLIN1 plays an important role in chicken lipid metabolism. To further reveal the role of PLIN1 in the growth and development of adipocytes, a chicken preadipocyte line with a PLIN1 gene knockout was established by the CRISPR/Cas9 gene editing technique, and the effects of the PLIN1 gene on the proliferation, apoptosis, differentiation and lipolysis of chicken preadipocytes were detected. The results showed that the CRISPR/Cas9 system effectively mediated knockout of the PLIN1 gene. After the deletion of PLIN1, the differentiation ability and early apoptotic activity of chicken preadipocytes decreased, and their proliferation ability increased. Moreover, knockout of PLIN1 promoted chicken preadipocyte lipolysis under basal conditions and inhibited chicken preadipocyte lipolysis under hormone stimulation. Taken together, our results inferred that PLIN1 plays a regulatory role in the process of proliferation, apoptosis, differentiation and lipolysis of chicken preadipocytes.
Collapse
Affiliation(s)
- Guiying Zhai
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yongjia Pang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yichong Zou
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xinyu Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Jie Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qi Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Zhiping Cao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ning Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yuxiang Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
8
|
Boeriu E, Boc AG, Borda A, Negrean RA, Feciche B, Boeriu AI, Horhat FG, Mot IC, Horhat ID, Ravulapalli M, Sabuni O, Adi A, Anjary A, Arghirescu ST. Insights on Lipomatosis after Platinum-Based Chemotherapy Use in Pediatric Oncology: A Case Report. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121715. [PMID: 36556917 PMCID: PMC9784424 DOI: 10.3390/medicina58121715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Agents of platinum-based chemotherapy, such as cisplatin or carboplatin, are used in the treatment of a wide range of malignancies that affect children, such as brain tumors, osteosarcoma, neuroblastoma, hepatoblastoma, and germ cell tumors (GCTs). The Cyclophosphamide Equivalent Dose (CED) calculator for reproductive risk does not take platinum-based chemotherapy into account, despite the fact that it accounts for the majority of chemotherapy medications that are typically administered for pediatric GCTs. As a result, exposure to platinum-based drugs throughout infancy can have predictable long-term effects such as infertility, as well as other rare encounters such as lipoma formation and lipomatosis. Lipomas are the most prevalent benign soft tissue tumor subtype. They may be either solitary entities or engaged in multiple lipomatosis, which may have a familial origin or be an acquired disorder. Chemotherapy is a possible cause of lipomatosis. Chemotherapy based on cisplatin has been linked to a variety of long-term consequences, including kidney damage, neurotoxicity, and pulmonary toxicity, and may even create secondary cancers. However, lipoma development is known to occur in fewer than 1 in 100 individuals, and only a few examples of multiple cutaneous lipomatosis triggered by this therapy have been documented. Here we present a very rare case of lipomatosis in a pediatric patient with GCT under cisplatin therapy, which might be the third report of this kind affecting children.
Collapse
Affiliation(s)
- Estera Boeriu
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Oncology and Haematology, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Alexandra Georgiana Boc
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Alexandra Borda
- Department of Oncology and Haematology, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | | | - Bogdan Feciche
- Department of Urology, Satu-Mare County Emergency Hospital, Strada Ravensburg 2, 440192 Satu-Mare, Romania
| | - Amalia Iulia Boeriu
- Klinikum Landshut, Teaching Hospital of the LMU Munich, Clinic for Anaesthesiology, Intensive Care Medicine and Pain Therapy, Robert-Koch-Strasse, 184034 Landshut, Germany
| | - Florin George Horhat
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Correspondence:
| | - Ion Cristian Mot
- Department of Ear-Nose-Throat, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ioana Delia Horhat
- Department of Ear-Nose-Throat, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | | | - Omar Sabuni
- Faculty of General Medicine, Altinbas University, Dilmenler Cd., 34217 Istanbul, Turkey
| | - Abduljabar Adi
- Faculty of General Medicine, Baskent University, Fatih Sultan, 06790 Ankara, Turkey
| | - Adnan Anjary
- Faculty of General Medicine, Yeditepe University, Kayısdagı Cd., 34755 Istanbul, Turkey
| | - Smaranda Teodora Arghirescu
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Oncology and Haematology, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| |
Collapse
|
9
|
Henry A, Gordillo-Marañón M, Finan C, Schmidt AF, Ferreira JP, Karra R, Sundström J, Lind L, Ärnlöv J, Zannad F, Mälarstig A, Hingorani AD, Lumbers RT. Therapeutic Targets for Heart Failure Identified Using Proteomics and Mendelian Randomization. Circulation 2022; 145:1205-1217. [PMID: 35300523 PMCID: PMC9010023 DOI: 10.1161/circulationaha.121.056663] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart failure (HF) is a highly prevalent disorder for which disease mechanisms are incompletely understood. The discovery of disease-associated proteins with causal genetic evidence provides an opportunity to identify new therapeutic targets. METHODS We investigated the observational and causal associations of 90 cardiovascular proteins, which were measured using affinity-based proteomic assays. First, we estimated the associations of 90 cardiovascular proteins with incident heart failure by means of a fixed-effect meta-analysis of 4 population-based studies, composed of a total of 3019 participants with 732 HF events. The causal effects of HF-associated proteins were then investigated by Mendelian randomization, using cis-protein quantitative loci genetic instruments identified from genomewide association studies in more than 30 000 individuals. To improve the precision of causal estimates, we implemented an Mendelian randomization model that accounted for linkage disequilibrium between instruments and tested the robustness of causal estimates through a multiverse sensitivity analysis that included up to 120 combinations of instrument selection parameters and Mendelian randomization models per protein. The druggability of candidate proteins was surveyed, and mechanism of action and potential on-target side effects were explored with cross-trait Mendelian randomization analysis. RESULTS Forty-four of ninety proteins were positively associated with risk of incident HF (P<6.0×10-4). Among these, 8 proteins had evidence of a causal association with HF that was robust to multiverse sensitivity analysis: higher CSF-1 (macrophage colony-stimulating factor 1), Gal-3 (galectin-3) and KIM-1 (kidney injury molecule 1) were positively associated with risk of HF, whereas higher ADM (adrenomedullin), CHI3L1 (chitinase-3-like protein 1), CTSL1 (cathepsin L1), FGF-23 (fibroblast growth factor 23), and MMP-12 (matrix metalloproteinase-12) were protective. Therapeutics targeting ADM and Gal-3 are currently under evaluation in clinical trials, and all the remaining proteins were considered druggable, except KIM-1. CONCLUSIONS We identified 44 circulating proteins that were associated with incident HF, of which 8 showed evidence of a causal relationship and 7 were druggable, including adrenomedullin, which represents a particularly promising drug target. Our approach demonstrates a tractable roadmap for the triangulation of population genomic and proteomic data for the prioritization of therapeutic targets for complex human diseases.
Collapse
Affiliation(s)
- Albert Henry
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
| | - María Gordillo-Marañón
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
| | - Chris Finan
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
| | - Amand F. Schmidt
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
| | - João Pedro Ferreira
- Unidade de Investigação e Desenvolvimento Cardiovascular, Rede de Investigação em Saúde, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Portugal (J.P.F.)
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
| | - Ravi Karra
- Division of Cardiology, Department of Medicine (R.K.), Duke University Medical Center, Durham, NC
- Department of Pathology (R.K.), Duke University Medical Center, Durham, NC
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.Ä.)
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (J.Ä.)
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna‚ Sweden (A.M.)
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, MA (A.M.)
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
| | - R. Thomas Lumbers
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
- Health Data Research UK London (R.T.L.), University College London, United Kingdom
| | - HERMES and SCALLOP Consortia
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
- Health Data Research UK London (R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
- Unidade de Investigação e Desenvolvimento Cardiovascular, Rede de Investigação em Saúde, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Portugal (J.P.F.)
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
- Division of Cardiology, Department of Medicine (R.K.), Duke University Medical Center, Durham, NC
- Department of Pathology (R.K.), Duke University Medical Center, Durham, NC
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.Ä.)
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (J.Ä.)
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna‚ Sweden (A.M.)
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, MA (A.M.)
| |
Collapse
|
10
|
Ponce-de-Leon M, Linseisen J, Peters A, Linkohr B, Heier M, Grallert H, Schöttker B, Trares K, Bhardwaj M, Gào X, Brenner H, Kamiński KA, Paniczko M, Kowalska I, Baumeister SE, Meisinger C. Novel associations between inflammation-related proteins and adiposity: A targeted proteomics approach across four population-based studies. Transl Res 2022; 242:93-104. [PMID: 34780968 DOI: 10.1016/j.trsl.2021.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 11/27/2022]
Abstract
Chronic low-grade inflammation has been proposed as a linking mechanism between obesity and the development of inflammation-related conditions such as insulin resistance and cardiovascular disease. Despite major advances in the last 2 decades, the complex relationship between inflammation and obesity remains poorly understood. Therefore, we aimed to identify novel inflammation-related proteins associated with adiposity. We investigated the association between BMI and waist circumference and 72 circulating inflammation-related proteins, measured using the Proximity Extension Assay (Olink Proteomics), in 3,308 participants of four independent European population-based studies (KORA-Fit, BVSII, ESTHER, and Bialystok PLUS). In addition, we used body fat mass measurements obtained by Dual-energy X-ray absorptiometry (DXA) in the Bialystok PLUS study to further validate our results and to explore the relationship between inflammation-related proteins and body fat distribution. We found 14 proteins associated with at least one measure of adiposity across all four studies, including four proteins for which the association is novel: DNER, SLAMF1, RANKL, and CSF-1. We confirmed previously reported associations with CCL19, CCL28, FGF-21, HGF, IL-10RB, IL-18, IL-18R1, IL-6, SCF, and VEGF-A. The majority of the identified inflammation-related proteins were associated with visceral fat as well as with the accumulation of adipose tissue in the abdomen and the trunk. In conclusion, our study provides new insights into the immune dysregulation observed in obesity that might help uncover pathophysiological mechanisms of disease development.
Collapse
Affiliation(s)
- Mariana Ponce-de-Leon
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Jakob Linseisen
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Birgit Linkohr
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany; KORA Study Centre, University Hospital Augsburg, Augsburg, Germany
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Kira Trares
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Megha Bhardwaj
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Xīn Gào
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Herman Brenner
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Karol Adam Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Marlena Paniczko
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | | | - Christa Meisinger
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
11
|
Molina-Ayala MA, Rodríguez-Amador V, Suárez-Sánchez R, León-Solís L, Gómez-Zamudio J, Mendoza-Zubieta V, Cruz M, Suárez-Sánchez F. Expression of obesity- and type-2 diabetes-associated genes in omental adipose tissue of individuals with obesity. Gene X 2022; 815:146181. [PMID: 34995730 DOI: 10.1016/j.gene.2021.146181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 12/19/2022] Open
Abstract
AIMS Obesity and type 2 diabetes mellitus are two pathologies that share metabolic abnormalities in most of the cases; however, there are differences as well. Some studies have reported that approximately 30% of obese patients have normal glucose and lipid levels in blood despite an accumulation of abdominal adipose tissue. Here, we compare the gene expression in adipose tissue of several genes associated with obesity and/or diabetes between obese patients without T2D and obese patients with T2D. METHODS Omental adipose tissue was collected during the patients elective bariatric surgery. Gene expression was determined by real-time PCR. Phenotypic variables were correlated with gene expression and 2^-ΔΔCt relative expression analysis between groups was performed. RESULTS The stronger correlations in the obese without T2D or reference group was between ICAM1 and HbA1c; HP and TC and LDL while in the obese with diabetes or case group the correlation occurred between CSF1 and BMI. A correlation between HP and TC was found in the case group as well. The expression of VEGFA, CCND2, IL1R1 and PTEN was downregulated in the obese with T2D group. CONCLUSIONS This study identified genes whose expression is different between obese subjects with and without diabetes. Those genes are related to inflammation, cholesterol transport, adipocyte differentiation/expansion and browning.
Collapse
Affiliation(s)
- Mario A Molina-Ayala
- Diabetes and Obesity Clinic, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico
| | - Virginia Rodríguez-Amador
- Medical and Biochemistry Research Unit, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico
| | - Rocío Suárez-Sánchez
- Laboratory of Genomic Medicine, 6th floor, CENIAQ, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Lizbel León-Solís
- Department of Microbiology, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Jaime Gómez-Zamudio
- Medical and Biochemistry Research Unit, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico
| | - Victoria Mendoza-Zubieta
- Endocrinology Unit, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico
| | - Miguel Cruz
- Medical and Biochemistry Research Unit, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico
| | - Fernando Suárez-Sánchez
- Medical and Biochemistry Research Unit, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, IMSS. Av. Cuauhtémoc 330, CP 06720 Mexico City, Mexico.
| |
Collapse
|
12
|
Freuchet A, Salama A, Remy S, Guillonneau C, Anegon I. IL-34 and CSF-1, deciphering similarities and differences at steady state and in diseases. J Leukoc Biol 2021; 110:771-796. [PMID: 33600012 DOI: 10.1002/jlb.3ru1120-773r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Although IL-34 and CSF-1 share actions as key mediators of monocytes/macrophages survival and differentiation, they also display differences that should be identified to better define their respective roles in health and diseases. IL-34 displays low sequence homology with CSF-1 but has a similar general structure and they both bind to a common receptor CSF-1R, although binding and subsequent intracellular signaling shows differences. CSF-1R expression has been until now mainly described at a steady state in monocytes/macrophages and myeloid dendritic cells, as well as in some cancers. IL-34 has also 2 other receptors, protein-tyrosine phosphatase zeta (PTPζ) and CD138 (Syndecan-1), expressed in some epithelium, cells of the central nervous system (CNS), as well as in numerous cancers. While most, if not all, of CSF-1 actions are mediated through monocyte/macrophages, IL-34 has also other potential actions through PTPζ and CD138. Additionally, IL-34 and CSF-1 are produced by different cells in different tissues. This review describes and discusses similarities and differences between IL-34 and CSF-1 at steady state and in pathological situations and identifies possible ways to target IL-34, CSF-1, and its receptors.
Collapse
Affiliation(s)
- Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Apolline Salama
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Remy
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
13
|
Generation of immune cell containing adipose organoids for in vitro analysis of immune metabolism. Sci Rep 2020; 10:21104. [PMID: 33273595 PMCID: PMC7713299 DOI: 10.1038/s41598-020-78015-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is an organized endocrine organ with important metabolic and immunological functions and immune cell-adipocyte crosstalk is known to drive various disease pathologies. Suitable 3D adipose tissue organoid models often lack resident immune cell populations and therefore require the addition of immune cells isolated from other organs. We have created the first 3D adipose tissue organoid model which could contain and maintain resident immune cell populations of the stromal vascular fraction (SVF) and proved to be effective in studying adipose tissue biology in a convenient manner. Macrophage and mast cell populations were successfully confirmed within our organoid model and were maintained in culture without the addition of growth factors. We demonstrated the suitability of our model for monitoring the lipidome during adipocyte differentiation in vitro and confirmed that this model reflects the physiological lipidome better than standard 2D cultures. In addition, we applied mass spectrometry-based lipidomics to track lipidomic changes in the lipidome upon dietary and immunomodulatory interventions. We conclude that this model represents a valuable tool for immune-metabolic research.
Collapse
|
14
|
Bray GA, Bouchard C. The biology of human overfeeding: A systematic review. Obes Rev 2020; 21:e13040. [PMID: 32515127 DOI: 10.1111/obr.13040] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/18/2020] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
This systematic review has examined more than 300 original papers dealing with the biology of overfeeding. Studies have varied from 1 day to 6 months. Overfeeding produced weight gain in adolescents, adult men and women and in older men. In longer term studies, there was a clear and highly significant relationship between energy ingested and weight gain and fat storage with limited individual differences. There is some evidence for a contribution of a genetic component to this response variability. The response to overfeeding was affected by the baseline state of the groups being compared: those with insulin resistance versus insulin sensitivity; those prone to obesity versus those resistant to obesity; and those with metabolically abnormal obesity versus those with metabolically normal obesity. Dietary components, such as total fat, polyunsaturated fat and carbohydrate influenced the patterns of adipose tissue distribution as did the history of low or normal birth weight. Overfeeding affected the endocrine system with increased circulating concentrations of insulin and triiodothyronine frequently present. Growth hormone, in contrast, was rapidly suppressed. Changes in plasma lipids were influenced by diet, exercise and the magnitude of weight gain. Adipose tissue and skeletal muscle morphology and metabolism are substantially altered by chronic overfeeding.
Collapse
Affiliation(s)
- George A Bray
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Claude Bouchard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| |
Collapse
|
15
|
Mukherjee S, Aseer KR, Yun JW. Roles of Macrophage Colony Stimulating Factor in White and Brown Adipocytes. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-020-0023-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Trindade R, Albrektsson T, Galli S, Prgomet Z, Tengvall P, Wennerberg A. Bone Immune Response to Materials, Part II: Copper and Polyetheretherketone (PEEK) Compared to Titanium at 10 and 28 Days in Rabbit Tibia. J Clin Med 2019; 8:jcm8060814. [PMID: 31181635 PMCID: PMC6616385 DOI: 10.3390/jcm8060814] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
Abstract
Osseointegration is likely the result of an immunologically driven bone reaction to materials such as titanium. Osseointegration has resulted in the clinical possibility to anchor oral implants in jaw bone tissue. However, the mechanisms behind bony anchorage are not fully understood and complications over a longer period of time have been reported. The current study aims at exploring possible differences between copper (Cu) and polyetheretherketone (PEEK) materials that do not osseointegrate, with osseointegrating cp titanium as control. The implants were placed in rabbit tibia and selected immune markers were evaluated at 10 and 28 days of follow-up. Cu and PEEK demonstrated at both time points a higher immune activation than cp titanium. Cu demonstrated distance osteogenesis due to a maintained proinflammatory environment over time, and PEEK failed to osseointegrate due to an immunologically defined preferential adipose tissue formation on its surface. The here presented results suggest the description of two different mechanisms for failed osseointegration, both of which are correlated to the immune system.
Collapse
Affiliation(s)
- Ricardo Trindade
- Department of Prosthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Tomas Albrektsson
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Silvia Galli
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Zdenka Prgomet
- Department of Oral Pathology, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Pentti Tengvall
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Ann Wennerberg
- Department of Prosthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
17
|
Lee SW, Kang NH, Choi JW. Functional Secretion of Granulocyte Colony Stimulating Factor in Bacillus subtilis and Its Thermogenic Activity in Brown Adipocytes. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0127-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
18
|
Mao R, Kurada S, Gordon IO, Baker ME, Gandhi N, McDonald C, Coffey JC, Rieder F. The Mesenteric Fat and Intestinal Muscle Interface: Creeping Fat Influencing Stricture Formation in Crohn's Disease. Inflamm Bowel Dis 2019; 25:421-426. [PMID: 30346528 DOI: 10.1093/ibd/izy331] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Indexed: 12/12/2022]
Abstract
Adipose tissue is present in close proximity to various organs in the human body. One prominent example is fat contained in the mesentery that is contiguous with all abdominal digestive organs including the intestine. Despite the fact that mesenteric fat-wrapping around the inflamed gut (so called "creeping fat") was described as a characteristic feature of Crohn's disease (CD) in the early 1930s, the functional implications of creeping fat have received only recent attention. As a potent producer of fatty acids, cytokines, growth factors, and adipokines, creeping fat plays an important role in regulation of immunity and inflammation. Increasing evidence points to a link between creeping fat and intestinal inflammation in CD, where histopathologic evaluation shows a significant association between creeping fat and connective tissue changes in the bowel wall, such as muscular hypertrophy, fibrosis, and stricture formation. In addition, emerging mechanistic data indicate a link between creeping fat, muscularis propria hyperplasia, and stricturing disease. Information on fat-mesenchymal interactions in other organs could provide clues to fill the fundamental knowledge gap on the role of distinct components of creeping fat in intestinal fibrosis and stricture formation. Future studies will provide important new information that in turn could lead to novel therapeutic agents aimed at prevention or treatment of CD-associated fibrosis and stricture formation.
Collapse
Affiliation(s)
- Ren Mao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, China.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute.,Department of Inflammation and Immunity, Lerner Research Institute
| | - Satya Kurada
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute.,Department of Inflammation and Immunity, Lerner Research Institute
| | - Ilyssa O Gordon
- Department of Pathology, Robert J. Tomsich Pathology & Laboratory Medicine Institute
| | - Mark E Baker
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute.,Section of Abdominal Imaging, Imaging Institute, The Cleveland Clinic Foundation, Cleveland, Ohio
| | - Namita Gandhi
- Section of Abdominal Imaging, Imaging Institute, The Cleveland Clinic Foundation, Cleveland, Ohio
| | | | - J Calvin Coffey
- Department of Surgery, Graduate Entry Medical School, University Hospital Limerick Group, Centre for Interventions in Infection, Inflammation and Immunity, University of Limerick, Limerick, Ireland
| | - Florian Rieder
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute.,Department of Inflammation and Immunity, Lerner Research Institute
| |
Collapse
|
19
|
Daryabor G, Kabelitz D, Kalantar K. An update on immune dysregulation in obesity-related insulin resistance. Scand J Immunol 2019; 89:e12747. [PMID: 30593678 DOI: 10.1111/sji.12747] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of the adipose tissue (AT) that might develop into systemic inflammation, insulin resistance (IR) and an increased risk of type 2 diabetes mellitus (T2DM) in severe obese rodents and humans. In the lean state, small normal adipocytes and AT macrophages interact with each other to maintain metabolic homeostasis but during obesity, enlarged adipocytes secrete inflammatory mediators and express immune receptors to recruit immune cells and aggravate the inflammation. The better understanding of the obesity-related inflammatory milieu and the sequential events leading to IR could be helpful in designing new preventive and therapeutic strategies. The present review will discuss the cellular and molecular abnormalities participating in the pathogenesis of obesity in obese individuals as well as high-fat diet (HFD)-fed mice, a mouse model of obesity.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kurosh Kalantar
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Chang CC, Sia KC, Chang JF, Lin CM, Yang CM, Huang KY, Lin WN. Lipopolysaccharide promoted proliferation and adipogenesis of preadipocytes through JAK/STAT and AMPK-regulated cPLA2 expression. Int J Med Sci 2019; 16:167-179. [PMID: 30662340 PMCID: PMC6332489 DOI: 10.7150/ijms.24068] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/04/2018] [Indexed: 12/15/2022] Open
Abstract
The proliferation and adipogenesis of preadipocytes played important roles in the development of adipose tissue and contributed much to the processes of obesity. On the other hand, lipopolysaccharide (LPS), also known as endotoxin, is a key outer membrane component of gram-negative bacteria in the gut microbiota, and has a dominant role in linking inflammation to high-fat diet-induced metabolic syndrome. Studies suggested the potential roles of LPS in hepatic steatosis and in obese mice models. However, the molecular mechanisms underlying LPS-regulated obesity remained largely unknown. Here we reported that LPS stimulated expression of cyosolic phospholipase A2 (cPLA2), one of inflammation regulators of obesity, in the preadipocytes. Pretreatment the inhibitors of JAK2, STAT3, STAT5 or AMPK significantly reduced LPS-increased mRNA and protein expression of cPLA2 together with phosphorylation of JAK2, STAT3, STAT5 and AMPK, separately. Similarly, transfection of siRNA against JAK2 or AMPK abolished expression of cPLA2 and phosphorylation of JAK2 or AMPK together with downregulated expression of JAK2 and AMPK protein. LPS enhanced activation of STAT3 and STAT5 via JAK2-dependent manner in the preadipocytes. Transfection of JAK2 or AMPK siRNA further proofed the independence of JAK2 and AMPK in LPS-treated preadipocytes. In addition, LPS-increased DNA synthesis, cell numbers and cell viability of preadipocytes were attenuated by AACOCF3, AG490, BML-275, cPLA2 siRNA, JAK2 siRNA or AMPK siRNA. Attenuation JAK2/STAT or AMPK-dependent cPLA2 expression reduced LPS-mediated adipogenesis of preadipocytes. Stimulation of arachidonic acid or AMPK activator, A-769662, increased cell numbers and cell viability and promoted differentiation of preadipocytes. Collectively, these results indicated that LPS increased preadipocytes proliferation and adipogenesis via JAK/STAT and AMPK-dependent cPLA2 expression. The mechanisms of LPS-stimulated cPLA2 expression may be a link between bacteria and obesity and provides the molecular basis for preventing metabolic syndrome or hyperplasic obesity.
Collapse
Affiliation(s)
- Chao-Chien Chang
- Division of Cardiology, Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pharmacology, School of medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Kee-Chin Sia
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Jia-Feng Chang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan.,PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Internal Medicine, En-Chu-Kong Hospital, New Taipei City, Taiwan
| | - Chia-Mo Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Chemistry, Fu-Jen Catholic University, New Taipei, Taiwan.,Division of Chest Medicine, Shin Kong Hospital, Taipei, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.,Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.,Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
21
|
Lovric A, Granér M, Bjornson E, Arif M, Benfeitas R, Nyman K, Ståhlman M, Pentikäinen MO, Lundbom J, Hakkarainen A, Sirén R, Nieminen MS, Lundbom N, Lauerma K, Taskinen MR, Mardinoglu A, Boren J. Characterization of different fat depots in NAFLD using inflammation-associated proteome, lipidome and metabolome. Sci Rep 2018; 8:14200. [PMID: 30242179 PMCID: PMC6155005 DOI: 10.1038/s41598-018-31865-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is recognized as a liver manifestation of metabolic syndrome, accompanied with excessive fat accumulation in the liver and other vital organs. Ectopic fat accumulation was previously associated with negative effects at the systemic and local level in the human body. Thus, we aimed to identify and assess the predictive capability of novel potential metabolic biomarkers for ectopic fat depots in non-diabetic men with NAFLD, using the inflammation-associated proteome, lipidome and metabolome. Myocardial and hepatic triglycerides were measured with magnetic spectroscopy while function of left ventricle, pericardial and epicardial fat, subcutaneous and visceral adipose tissue were measured with magnetic resonance imaging. Measured ectopic fat depots were profiled and predicted using a Random Forest algorithm, and by estimating the Area Under the Receiver Operating Characteristic curves. We have identified distinct metabolic signatures of fat depots in the liver (TAG50:1, glutamate, diSM18:0 and CE20:3), pericardium (N-palmitoyl-sphinganine, HGF, diSM18:0, glutamate, and TNFSF14), epicardium (sphingomyelin, CE20:3, PC38:3 and TNFSF14), and myocardium (CE20:3, LAPTGF-β1, glutamate and glucose). Our analyses highlighted non-invasive biomarkers that accurately predict ectopic fat depots, and reflect their distinct metabolic signatures in subjects with NAFLD.
Collapse
Affiliation(s)
- Alen Lovric
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Marit Granér
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Elias Bjornson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Rui Benfeitas
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Kristofer Nyman
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Markku O Pentikäinen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jesper Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Reijo Sirén
- Department of General Practice and Primary Health Care, Health Care Centre of City of Helsinki and University of Helsinki, Helsinki, Finland
| | - Markku S Nieminen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Nina Lundbom
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Kirsi Lauerma
- Department of Radiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Marja-Riitta Taskinen
- Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden. .,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Jan Boren
- Department of Molecular and Clinical Medicine/Wallenberg Lab, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
22
|
Xiong Y, Russell DL, McDonald LT, Cowart LA, LaRue AC. Hematopoietic Stem Cell-derived Adipocytes Promote Tumor Growth and Cancer Cell Migration. ACTA ACUST UNITED AC 2017; 3. [PMID: 28989976 PMCID: PMC5627654 DOI: 10.16966/2381-3318.130] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Adipocytes, apart from their critical role as the energy storage depots, contribute to the composition of the tumor microenvironment. Our previous studies based on a single hematopoietic stem cell (HSC) transplantation model, have revealed a novel source of adipocytes from HSCs via monocyte/macrophage progenitors. Herein, we extend these studies to examine the role of HSC-derived adipocytes (HSC-Ad) in tumor progression. When cultured under adipogenic conditions, bone marrow-derived monocytic progenitors differentiated into adipocytes that accumulated oil droplets containing triglyceride. The adipokine array and ELISAs confirmed secretion of multiple adipokines by HSC-Ad. These adipocytes underwent further development in vivo when injected subcutaneously into C57Bl/6 mice. When co-injected with melanoma B16F1 cells or breast cancer E0771 cells into syngeneic C57Bl/6 mice, HSC-Ad not only accelerated both melanoma and breast tumor growth, but also enhanced vascularization in both tumors. Conditioned media from HSC-Ad supported B16F1 and E0771 cell proliferation and enhanced cell migration in vitro. Among the HSC-Ad secreted adipokines, insulin-like growth factor 1 (IGF-1) played an important role in E0771 cell proliferation. Hepatocyte growth factor (HGF) was indispensable for B16F1 cell migration, whereas HGF and platelet-derived growth factor BB (PDGF-BB) collectively contributed to E0771 cell migration. Expression levels of receptors for IGF-1, HGF, and PDGF-BB correlated with their differential roles in B16F1 and E0771 cell proliferation and migration. Our data suggest that HSC-Ad differentially regulate tumor behavior through distinct mechanisms.
Collapse
Affiliation(s)
- Y Xiong
- Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - D L Russell
- Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - L T McDonald
- Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - L A Cowart
- Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA.,Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - A C LaRue
- Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
23
|
Serena C, Keiran N, Ceperuelo-Mallafre V, Ejarque M, Fradera R, Roche K, Nuñez-Roa C, Vendrell J, Fernández-Veledo S. Obesity and Type 2 Diabetes Alters the Immune Properties of Human Adipose Derived Stem Cells. Stem Cells 2016; 34:2559-2573. [DOI: 10.1002/stem.2429] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/18/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Carolina Serena
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Noelia Keiran
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Victoria Ceperuelo-Mallafre
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Miriam Ejarque
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | | | - Kelly Roche
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Catalina Nuñez-Roa
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Joan Vendrell
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Sonia Fernández-Veledo
- Hospital Universitari De Tarragona Joan XXIII, Institut D´Investigació Sanitària Pere Virgili; Universitat Rovira I Virgili; Tarragona Spain
- Instituto De Salud Carlos III; CIBER De Diabetes Y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| |
Collapse
|
24
|
Devêvre EF, Renovato-Martins M, Clément K, Sautès-Fridman C, Cremer I, Poitou C. Profiling of the three circulating monocyte subpopulations in human obesity. THE JOURNAL OF IMMUNOLOGY 2015; 194:3917-23. [PMID: 25786686 DOI: 10.4049/jimmunol.1402655] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/04/2015] [Indexed: 12/24/2022]
Abstract
Three subpopulations of circulating monocytes have been described: CD14(2+)CD16(-) (classical monocytes [CM]), CD14(2+)CD16(+) (intermediate monocytes [IM]), and CD14(+)CD16(2+) (nonclassical monocytes [NCM]). We previously showed that obesity is associated with an increased proportion of IM and NCM. Our objective is to decipher the migratory and inflammatory functions of each monocyte subset in obesity-related low-grade inflammation. Twenty-six healthy, normal-weight and nondiabetic volunteers (C) and 40 obese nondiabetic (Ob) individuals were included in this study. We explored the gene expression profile of 18 inflammatory genes in each subset of C and Ob subjects and measured protein expression of the upregulated genes. We then tested their functional response to TLR signaling in both groups. We showed an increased expression of CX3CR1 in all monocyte subpopulations and of CCR2 and CCR5 in CM and IM in the Ob group. We found negative correlation between CCR2 and CX3CR1 expressions and high-density lipoprotein-cholesterol, whereas CCR5 expression was positively linked to obesity-related metabolic traits. Production of inflammatory proteins upon bacterial LPS and viral ssRNA stimulation was higher in CM and NCM of the Ob group compared with the C group. Our work highlights an enhanced inflammatory phenotype of monocytes with a higher response to TLR4 and TLR8 stimulations in obesity. Moreover, it suggests an increased migration capacity of CM and IM subpopulations.
Collapse
Affiliation(s)
- Estelle F Devêvre
- INSERM, Unité Mixte de Recherche 1138, Team 13, Centre de Recherches des Cordeliers, F-75006 Paris, France; INSERM, U1166, Nutriomic Team 6, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France; Centre d'Imagerie Cellulaire et de Cytométrie, Centre de Recherche des Cordeliers, Unité Mixte de Recherche 1138, F-75006 Paris, France
| | - Mariana Renovato-Martins
- INSERM, Unité Mixte de Recherche 1138, Team 13, Centre de Recherches des Cordeliers, F-75006 Paris, France; INSERM, U1166, Nutriomic Team 6, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France
| | - Karine Clément
- INSERM, U1166, Nutriomic Team 6, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France; Institute of Cardiometabolism and Nutrition, F-75013 Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Département Nutrition, F-75013 Paris, France; and Centre de Recherche en Nutrition Humaine-Ile de France, F-75013 Paris, France
| | - Catherine Sautès-Fridman
- INSERM, Unité Mixte de Recherche 1138, Team 13, Centre de Recherches des Cordeliers, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France
| | - Isabelle Cremer
- INSERM, Unité Mixte de Recherche 1138, Team 13, Centre de Recherches des Cordeliers, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France;
| | - Christine Poitou
- INSERM, U1166, Nutriomic Team 6, F-75006 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, F-75013 Paris, France; Université Paris Descartes, F-75006 Paris, France; Institute of Cardiometabolism and Nutrition, F-75013 Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Département Nutrition, F-75013 Paris, France; and Centre de Recherche en Nutrition Humaine-Ile de France, F-75013 Paris, France
| |
Collapse
|
25
|
Konenkov VI, Klimontov VV, Myakina NE, Tyan NV, Fazullina ON, Romanov VV. Increased serum concentrations of inflammatory cytokines in type 2 diabetic patients with chronic kidney disease. TERAPEVT ARKH 2015; 87:45-49. [DOI: 10.17116/terarkh201587645-49] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Sagayama H, Jikumaru Y, Hirata A, Yamada Y, Yoshimura E, Ichikawa M, Hatamoto Y, Ebine N, Kiyonaga A, Tanaka H, Higaki Y. Measurement of body composition in response to a short period of overfeeding. J Physiol Anthropol 2014; 33:29. [PMID: 25208693 PMCID: PMC4237876 DOI: 10.1186/1880-6805-33-29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/12/2014] [Indexed: 11/17/2022] Open
Abstract
Background Obesity and overweight are increasing in prevalence in developed countries as a result of changing dietary habits and a lack of physical activity. The purpose of the present study was to evaluate the changes in body composition during short-term overfeeding using the three-component model, which is composed of fat mass (FM), total body water (TBW), and fat-free dry solids (FFDS). Methods Ten healthy men completed 3 days of overfeeding during which they consumed 1,500 kcal/day more energy than consumed in their normal diets. Body composition was evaluated at three time points: the day before and after their normal diets and the day after the 3-day overfeeding diet. Results Before and after their normal diets, there were no significant differences in body weight and composition, but after 3 days of overfeeding, body weight, TBW, and FFDS increased 0.7, 0.7, and 0.2 kg, respectively (P <0.0001). There was no significant difference in FM between the normal and overfeeding diets. Conclusion This study suggests that TBW gain contributes to weight gain following a short-term overfeeding.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Yasuki Higaki
- Fukuoka University Institute for Physical Activity, 8-19-1 Nanakuma, Jounan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
27
|
Pires PW, Girgla SS, McClain JL, Kaminski NE, van Rooijen N, Dorrance AM. Improvement in middle cerebral artery structure and endothelial function in stroke-prone spontaneously hypertensive rats after macrophage depletion. Microcirculation 2014; 20:650-61. [PMID: 23647512 DOI: 10.1111/micc.12064] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 04/30/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inflammation is involved in the pathogenesis of hypertension. Hypertensive animals have an increased number of perivascular macrophages in cerebral arteries. Macrophages might be involved in remodeling of the cerebral vasculature. We hypothesized that peripheral macrophage depletion would improve MCA structure and function in hypertensive rats. METHODS For macrophage depletion, six-week-old stroke-prone spontaneously hypertensive rats (SHRSP) were treated with CLOD, 10 mL/kg every three or four days, i.p., or vehicle (PBS lipo). MCA structure and function were analyzed by pressure and wire myography. RESULTS Blood pressure was not affected by CLOD. The number of perivascular CD163-positive cells per microscopic field was reduced in the brain of SHRSP+CLOD. CLOD treatment caused an improvement in endothelium-dependent dilation after intralumenal perfusion of ADP and incubation with Ach. Inhibition of NO production blunted the Ach response, and endothelium-independent dilation was not altered. At an intralumenal pressure of 80 mmHg, MCA from SHRSP+CLOD showed increased lumen diameter, decreased wall thickness, and wall-to-lumen ratio. Cross-sectional area of pial arterioles from SHRSP+CLOD was higher than PBS lipo. CONCLUSIONS These results suggest that macrophage depletion attenuates MCA remodeling and improves MCA endothelial function in SHRSP.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | | | | | | | | | | |
Collapse
|
28
|
Kruis T, Batra A, Siegmund B. Bacterial translocation - impact on the adipocyte compartment. Front Immunol 2014; 4:510. [PMID: 24432024 PMCID: PMC3881001 DOI: 10.3389/fimmu.2013.00510] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/24/2013] [Indexed: 12/29/2022] Open
Abstract
Over the last decade it became broadly recognized that adipokines and thus the fat tissue compartment exert a regulatory function on the immune system. Our own group described the pro-inflammatory function of the adipokine leptin within intestinal inflammation in a variety of animal models. Following-up on this initial work, the aim was to reveal stimuli and mechanisms involved in the activation of the fat tissue compartment and the subsequent release of adipokines and other mediators paralleled by the infiltration of immune cells. This review will summarize the current literature on the possible role of the mesenteric fat tissue in intestinal inflammation with a focus on Crohn’s disease (CD). CD is of particular interest in this context since the transmural intestinal inflammation has been associated with a characteristic hypertrophy of the mesenteric fat, a phenomenon called “creeping fat.” The review will address three consecutive questions: (i) What is inducing adipocyte activation, (ii) which factors are released after activation and what are the consequences for the local fat tissue compartment and infiltrating cells; (iii) do the answers generated before allow for an explanation of the role of the mesenteric fat tissue within intestinal inflammation? With this review we will provide a working model indicating a close interaction in between bacterial translocation, activation of the adipocytes, and subsequent direction of the infiltrating immune cells. In summary, the models system mesenteric fat indicates a unique way how adipocytes can directly interact with the immune system.
Collapse
Affiliation(s)
- Tassilo Kruis
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Arvind Batra
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Britta Siegmund
- Department of Medicine I (Gastroenterology, Rheumatology, Infectious Diseases), Charité - Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
29
|
Abstract
Adipose tissue plays a major role in metabolic homeostasis, which it coordinates through a number of local and systemic effectors. The burgeoning epidemic of metabolic disease, especially obesity and type 2 diabetes, has focused attention on the adipocyte. In this chapter, we review strategies for genetic overexpression and knockout of specific genes in adipose tissue. We also discuss these strategies in the context of different types of adipocytes, including brown, beige, and white fat cells.
Collapse
Affiliation(s)
- Sona Kang
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Xingxing Kong
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Evan D Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
30
|
Lee YH, Mottillo EP, Granneman JG. Adipose tissue plasticity from WAT to BAT and in between. Biochim Biophys Acta Mol Basis Dis 2013; 1842:358-69. [PMID: 23688783 DOI: 10.1016/j.bbadis.2013.05.011] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/21/2013] [Accepted: 05/06/2013] [Indexed: 01/09/2023]
Abstract
Adipose tissue plays an essential role in regulating energy balance through its metabolic, cellular and endocrine functions. Adipose tissue has been historically classified into anabolic white adipose tissue and catabolic brown adipose tissue. An explosion of new data, however, points to the remarkable heterogeneity among the cells types that can become adipocytes, as well as the inherent metabolic plasticity of mature cells. These data indicate that targeting cellular and metabolic plasticity of adipose tissue might provide new avenues for treatment of obesity-related diseases. This review will discuss the developmental origins of adipose tissue, the cellular complexity of adipose tissues, and the identification of progenitors that contribute to adipogenesis throughout development. We will touch upon the pathological remodeling of adipose tissue and discuss how our understanding of adipose tissue remodeling can uncover new therapeutic targets. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Yun-Hee Lee
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Emilio P Mottillo
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - James G Granneman
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
31
|
Yachi R, Muto C, Ohtaka N, Aoki Y, Koike T, Igarashi O, Kiyose C. Effects of tocotrienol on tumor necrosis factor-α/d-galactosamine-induced steatohepatitis in rats. J Clin Biochem Nutr 2013; 52:146-53. [PMID: 23526264 PMCID: PMC3593132 DOI: 10.3164/jcbn.12-101] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/05/2012] [Indexed: 12/31/2022] Open
Abstract
It has been reported that α-tocopherol (α-Toc), a vitamin E analog, is effective for treatment of non-alcoholic steatohepatitis (NASH). However, it is unknown whether or not other vitamin E analogs are effective. Therefore we designed a new rat model of steatohepatitis induced by tumor necrosis factor-α (TNF-α) stimulation, and used it to investigate the effects of vitamin E analogs. The rat liver triglyceride content increased with the dosage of TNF-α/d-galactosamine (GalN), but was suppressed by intake of both tocotrienol (T3) and α-tocopherol. Moreover, lipid peroxides (thiobarbituric acid-reactive substances) level in the liver level was also lower in both groups after tocotrienol and α-Toc intake. Intake of both tocotrienol and α-tocopherol also tended to control the increase of liver damage marker activity. In the tocotrienol and α-tocopherol groups, increases of inflammatory cytokines mRNA expression in the liver were inhibited, and these effects were considered to contribute to improvement of inflammation and fibrosis. The expression of mRNAs for inflammatory cytokines in rat primary hepatocytes was increased by TNF-α stimulation, but was inhibited by addition of α-tocotrienol and γ-tocotrienol. Transforming growth factor-β1 mRNA expression in particular was significantly inhibited by γ-tocotrienol. These findings suggest that tocotrienol species are effective for amelioration of steatohepatitis, and that tocotrienol and α-tocopherol exert a synergistic effect.
Collapse
Affiliation(s)
- Rieko Yachi
- Department of Applied Chemistry, Graduate School of Kanagawa Institute of Technology, Kanagawa 243-0292, Japan ; Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Chang Q, Lu F. A novel strategy for creating a large amount of engineered fat tissue with an axial vascular pedicle and a prefabricated scaffold. Med Hypotheses 2012; 79:267-70. [PMID: 22688400 DOI: 10.1016/j.mehy.2012.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 04/09/2012] [Accepted: 05/02/2012] [Indexed: 01/06/2023]
Abstract
In plastic and reconstructive surgery, there is a tremendous clinical need for adequate implants are needed to restore soft-tissue defects resulting from tumor resection, traumatic injury, or congenital anomalies. Restoring the aesthetic function of the soft tissue is as important as restoring the natural tissue function. To address aesthetic issues, injection of hyaluronic acid and collagen and use of artificially synthesized biomaterials and autologous fat tissue grafts is extensive in the clinic, still faces limitations. Achieving minimal morbidity while compensating contour irregularities remains a major challenge because the available reconstruction methods and unsatisfactory biomaterials. Adipose tissue engineering holds great promise for reconstruction, but so far, there was no reports of large-volume engineered adipose tissue. Construction of a large volume of vascularized engineered fat tissue may overcome clinical challenges because vascularization is essential for the survival of engineered fat tissue and its integration with the host tissue. An arteriovenous bundle model for soft tissue has been used in prefabricating a large volume fat tissue with axial vascularization in vivo. Therefore, we hypothesized that combining adipose tissue-derived stem cells (ASCs), and prefabricated vascularized collagen scaffolds, with perforated chamber and arteriovenous bundle, could generate a large volume of engineered fat tissue with an axial vascular pedicle in vivo. Like vascularized autologous tissue, the new constructs could be transferred to the defective site by local transference or microsurgical techniques. The novel strategy could provide a large volume of engineered fat tissue suitable for clinical application and new therapeutic strategies for reconstructing defects if the hypothesis proved to be practical.
Collapse
Affiliation(s)
- Qiang Chang
- Department of Plastic and Cosmetic Surgery, Southern Hospital, Southern Medical University, Guangzhou North Road, 1838 Guangzhou, Guangdong 510515, PR China
| | | |
Collapse
|
33
|
Na HN, Nam JH. Adenovirus 36 as an obesity agent maintains the obesity state by increasing MCP-1 and inducing inflammation. J Infect Dis 2012; 205:914-22. [PMID: 22275403 DOI: 10.1093/infdis/jir864] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although it is well known that adenovirus 36 (Ad36) is associated with obesity in humans as well as in animals, the detailed cellular mechanism is unclear. METHODS Wild-type (WT) mice and monocyte chemoattractant protein-1 knockout (MCP-1(-/-)) mice were infected with Ad36, and their weights and inflammatory status were measured. Macrophage infiltration was examined in their reproductive fat pads and in a coculture system. The correlation between Ad36 antibody presence and MCP-1 levels was tested in human samples. RESULTS We have shown that Ad36 infection stimulated an inflammatory state by increasing the level of MCP-1 through the activation of nuclear factor κB, which in turn induced the infiltration of macrophages into adipocytes. This induced inflammation resulted in viral obesity, which caused chronic inflammation and affected lipid metabolism. In contrast to WT mice, MCP-1(-/-) mice were protected from Ad36-induced inflammation and obesity. The MCP-1 levels in Ad36-antibody-positive human group were higher than those in the antibody-negative group. CONCLUSIONS These findings support the proposition that virus-induced inflammation is the cellular mechanism underlying Ad36-induced obesity. These results also suggest that MCP-1 plays a critical role in Ad36-induced obesity and that MCP-1 may be a therapeutic target in preventing virus-induced obesity.
Collapse
Affiliation(s)
- Ha-Na Na
- Department of Biotechnology, The Catholic University of Korea, Wonmi-gu, Bucheon, Gyeonggi-do, Korea
| | | |
Collapse
|
34
|
Li H, Lee JH, Kim SY, Yun HY, Baek KJ, Kwon NS, Yoon Y, Jeong JH, Kim DS. Phosphatidylcholine induces apoptosis of 3T3-L1 adipocytes. J Biomed Sci 2011; 18:91. [PMID: 22145579 PMCID: PMC3261832 DOI: 10.1186/1423-0127-18-91] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/07/2011] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Phosphatidylcholine (PPC) formulation is used for lipolytic injection, even though its mechanism of action is not well understood. METHODS The viability of 3T3-L1 pre-adipocytes and differentiated 3T3-L1 cells was measured after treatment of PPC alone, its vehicle sodium deoxycholate (SD), and a PPC formulation. Western blot analysis was performed to examine PPC-induced signaling pathways. RESULTS PPC, SD, and PPC formulation significantly decreased 3T3-L1 cell viability in a concentration-dependent manner. PPC alone was not cytotoxic to CCD-25Sk human fibroblasts at concentrations <1 mg/ml, whereas SD and PPC formulation were cytotoxic. Western blot analysis demonstrated that PPC alone led to the phosphorylation of the stress signaling proteins, such as p38 mitogen-activated protein kinase and c-Jun N-terminal kinase, and activated caspase-9, -8, -3 as well as cleavage of poly(ADP-ribose) polymerase. However, SD did not activate the apoptotic pathways. Instead, SD and PPC formulation induced cell membrane lysis, which may lead to necrosis of cells. CONCLUSIONS PPC results in apoptosis of 3T3-L1 cells.
Collapse
Affiliation(s)
- Hailan Li
- Departments of Biochemistry, Chung-Ang University College of Medicine, Dongjak-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hofmann C, Chen N, Obermeier F, Paul G, Büchler C, Kopp A, Falk W, Schäffler A. C1q/TNF-related protein-3 (CTRP-3) is secreted by visceral adipose tissue and exerts antiinflammatory and antifibrotic effects in primary human colonic fibroblasts. Inflamm Bowel Dis 2011; 17:2462-71. [PMID: 21351204 DOI: 10.1002/ibd.21647] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 12/31/2010] [Indexed: 12/13/2022]
Abstract
BACKGROUND The adipokine CTRP-3 (C1q/TNF-related protein-3) belongs to the C1q/TNF-related protein family which antagonizes the effects of lipopolysaccharide (LPS). The aim was to investigate the antiinflammatory and antifibrotic role of CTRP-3 in Crohn's disease (CD). METHODS Mesenteric adipose tissue (MAT) of patients with CD or colonic cancer (CC) was resected. Human primary colonic lamina propria fibroblasts (CLPF) were isolated from controls and CD patients. Concentrations of chemokines and cytokines in the supernatants were measured by enzyme-linked immunosorbent assay (ELISA). Expression of connective tissue growth factor (CTGF), collagen I, and collagen III was analyzed by real-time polymerase chain reaction (PCR). Recombinant CTRP-3 expressed in insect cells was used for stimulation experiments. RESULTS CTRP-3 is synthesized and secreted by MAT resected from patients with CD, ulcerative colitis (UC), CC, and sigma diverticulitis as well as by murine and human mature adipocytes. CTRP-3 had no effect on the basal secretion of MCSF, MIF, or RANTES in MAT of CD and control patients. LPS-stimulation (10 ng/mL) significantly increased IL-8 release in CLPF of CD patients and, to a lesser extent, in cells of controls and of fibrotic CD tissue. CTRP-3 significantly and dose-dependently reduced LPS-induced IL-8 secretion in CLPF within 8 hours after LPS exposure, whereas LPS-induced IL-6 and TNF release was not affected. CTRP-3 inhibited TGF-β production and the expression of CTGF and collagen I in CLPF, whereas collagen III expression remained unchanged. CONCLUSIONS CTRP-3 exerts potent antiinflammatory and antifibrotic effects in CLPF by antagonizing the LPS pathway and by targeting the TGF-β-CTGF-collagen I pathway.
Collapse
Affiliation(s)
- Claudia Hofmann
- Department of Internal Medicine I, University Medical Center Regensburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Trudel G, Ryan SE, Rakhra K, Uhthoff HK. Extra- and Intramuscular Fat Accumulation Early after Rabbit Supraspinatus Tendon Division: Depiction with CT. Radiology 2010; 255:434-41. [DOI: 10.1148/radiol.10091377] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Massiera F, Barbry P, Guesnet P, Joly A, Luquet S, Moreilhon-Brest C, Mohsen-Kanson T, Amri EZ, Ailhaud G. A Western-like fat diet is sufficient to induce a gradual enhancement in fat mass over generations. J Lipid Res 2010; 51:2352-61. [PMID: 20410018 DOI: 10.1194/jlr.m006866] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The prevalence of obesity has steadily increased over the last few decades. During this time, populations of industrialized countries have been exposed to diets rich in fat with a high content of linoleic acid and a low content of alpha-linolenic acid compared with recommended intake. To assess the contribution of dietary fatty acids, male and female mice fed a high-fat diet (35% energy as fat, linoleic acid:alpha-linolenic acid ratio of 28) were mated randomly and maintained after breeding on the same diet for successive generations. Offspring showed, over four generations, a gradual enhancement in fat mass due to combined hyperplasia and hypertrophy with no change in food intake. Transgenerational alterations in adipokine levels were accompanied by hyperinsulinemia. Gene expression analyses of the stromal vascular fraction of adipose tissue, over generations, revealed discrete and steady changes in certain important players, such as CSF3 and Nocturnin. Thus, under conditions of genome stability and with no change in the regimen over four generations, we show that a Western-like fat diet induces a gradual fat mass enhancement, in accordance with the increasing prevalence of obesity observed in humans.
Collapse
Affiliation(s)
- Florence Massiera
- Université de Nice Sophia-Antipolis, CNRS, IBDC, UMR 6543, 06107 Nice, France
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ogawa M, Larue AC, Watson PM, Watson DK. Hematopoietic stem cell origin of connective tissues. Exp Hematol 2010; 38:540-7. [PMID: 20412832 DOI: 10.1016/j.exphem.2010.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 04/06/2010] [Accepted: 04/08/2010] [Indexed: 02/06/2023]
Abstract
Connective tissue consists of "connective tissue proper," which is further divided into loose and dense (fibrous) connective tissues and "specialized connective tissues." Specialized connective tissues consist of blood, adipose tissue, cartilage, and bone. In both loose and dense connective tissues, the principal cellular element is fibroblasts. It has been generally believed that all cellular elements of connective tissue, including fibroblasts, adipocytes, chondrocytes, and bone cells, are generated solely by mesenchymal stem cells. Recently, a number of studies, including those from our laboratory based on transplantation of single hematopoietic stem cells, strongly suggested a hematopoietic stem cell origin of these adult mesenchymal tissues. This review summarizes the experimental evidence for this new paradigm and discusses its translational implications.
Collapse
Affiliation(s)
- Makio Ogawa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC29401-5799, USA.
| | | | | | | |
Collapse
|
39
|
Hematopoietic stem cell origin of mesenchymal cells: opportunity for novel therapeutic approaches. Int J Hematol 2010; 91:353-9. [PMID: 20336396 DOI: 10.1007/s12185-010-0554-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 03/08/2010] [Indexed: 12/20/2022]
Abstract
There has been a general belief that there are two types of adult stem cells, i.e., hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs), each with distinctly different functions. According to this dogma, HSCs produce blood cells, while MSCs are thought to generate a number of non-hematopoietic cells including fibroblasts, adipocytes, chondrocytes and bone cells. Recently, a number of studies, including those in our laboratory based on single HSC transplantation, blurred the clear distinction between HSCs and MSCs and strongly suggested an HSC origin of the adult mesenchymal tissues. This review summarizes the experimental evidence for this new paradigm and the literature pointing out the vagary in the stem cell nature of MSCs. The concept of the HSC origin of mesenchymal cells will have many immediate and long-term impacts on the therapies of diseases and injuries of the connective tissues.
Collapse
|
40
|
McAllister EJ, Dhurandhar NV, Keith SW, Aronne LJ, Barger J, Baskin M, Benca RM, Biggio J, Boggiano MM, Eisenmann JC, Elobeid M, Fontaine KR, Gluckman P, Hanlon EC, Katzmarzyk P, Pietrobelli A, Redden DT, Ruden DM, Wang C, Waterland RA, Wright SM, Allison DB. Ten putative contributors to the obesity epidemic. Crit Rev Food Sci Nutr 2009; 49:868-913. [PMID: 19960394 PMCID: PMC2932668 DOI: 10.1080/10408390903372599] [Citation(s) in RCA: 449] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The obesity epidemic is a global issue and shows no signs of abating, while the cause of this epidemic remains unclear. Marketing practices of energy-dense foods and institutionally-driven declines in physical activity are the alleged perpetrators for the epidemic, despite a lack of solid evidence to demonstrate their causal role. While both may contribute to obesity, we call attention to their unquestioned dominance in program funding and public efforts to reduce obesity, and propose several alternative putative contributors that would benefit from equal consideration and attention. Evidence for microorganisms, epigenetics, increasing maternal age, greater fecundity among people with higher adiposity, assortative mating, sleep debt, endocrine disruptors, pharmaceutical iatrogenesis, reduction in variability of ambient temperatures, and intrauterine and intergenerational effects as contributing factors to the obesity epidemic are reviewed herein. While the evidence is strong for some contributors such as pharmaceutical-induced weight gain, it is still emerging for other reviewed factors. Considering the role of such putative etiological factors of obesity may lead to comprehensive, cause specific, and effective strategies for prevention and treatment of this global epidemic.
Collapse
Affiliation(s)
- Emily J McAllister
- Department of Infections and Obesity, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee J, Lee J, Hwang H, Jung E, Huh S, Hyun J, Park D. Promotion of stem cell proliferation by vegetable peptone. Cell Prolif 2009; 42:595-601. [PMID: 19614679 PMCID: PMC6496542 DOI: 10.1111/j.1365-2184.2009.00630.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 11/12/2008] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Technical limitations and evolution of therapeutic applications for cell culture-derived products have accelerated elimination of animal-derived constituents from such products to minimize inadvertent introduction of microbial contaminants, such as fungi, bacteria or viruses. The study described here was conducted to investigate the proliferative effect of vegetable peptone on adult stem cells in the absence of serum, and its possible mechanisms of action. MATERIALS AND METHODS Cell viability and proliferation were determined using the MTT assay and Click-iT EdU flow cytometry, respectively. In addition, changes in expression of cytokine genes were analysed using MILLIPLEX human cytokine enzyme-linked immunosorbent assay kit. RESULTS Viability of cord blood-derived mesenchymal stem cells (CB-MSC) and adipose tissue-derived stem cells (ADSC) increased significantly when treated with the peptone. In addition, median value of the group treated with peptone shifted to the right when compared to the untreated control group. Furthermore, quantitative analysis of the cytokines revealed that production of vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1), and interleukin-6 (IL-6) increased significantly in response to treatment with our vegetable peptone in both CB-MSCs and ADSCs. CONCLUSIONS Our findings revealed that the vegetable peptone promotes proliferation of CB-MSCs and ADSCs. In addition, results of this study suggest that induction of stem cell proliferation by vegetable peptone is likely to be related to its induction of VEGF, TGF-beta1, and IL-6 expression.
Collapse
Affiliation(s)
- J Lee
- Biospectrum Life Science Institute, Gunpo-City, Gyunggi-Do, South Korea
| | | | | | | | | | | | | |
Collapse
|
42
|
Sera Y, LaRue AC, Moussa O, Mehrotra M, Duncan JD, Williams CR, Nishimoto E, Schulte BA, Watson PM, Watson DK, Ogawa M. Hematopoietic stem cell origin of adipocytes. Exp Hematol 2009; 37:1108-20, 1120.e1-4. [PMID: 19576951 DOI: 10.1016/j.exphem.2009.06.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 06/26/2009] [Indexed: 12/20/2022]
Abstract
OBJECTIVE It has generally been believed that adipocytes are derived from mesenchymal stem cells via fibroblasts. We recently reported that fibroblasts/myofibroblasts in a number of tissues and organs are derived from hematopoietic stem cells (HSCs). In the present study, we tested the hypothesis that HSCs also give rise to adipocytes. MATERIALS AND METHODS Using transplantation of a single enhanced green fluorescent protein-positive (EGFP(+)) HSC and primary culture, we examined generation of adipocytes from HSCs. RESULTS Adipose tissues from clonally engrafted mice showed EGFP(+) adipocytes that stained positive for leptin, perilipin, and fatty acid binding protein 4. A diet containing rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, significantly enhanced the number of EGFP(+) adipocytes. When EGFP(+) bone marrow cells from clonally engrafted mice were cultured under adipogenic conditions, all of the cultured cells stained positive with Oil Red O and Sudan Black B and exhibited the presence of abundant mRNA for adipocyte markers. Finally, clonal culture- and sorting-based studies of Mac-1 expression of hematopoietic progenitors suggested that adipocytes are derived from HSCs via progenitors for monocytes/macrophages. CONCLUSION Together, these studies clarify the current controversy regarding the ability of HSCs to give rise to adipocytes. Furthermore, our primary culture method that generates adipocytes from uncommitted hematopoietic cells should contribute to the studies of the mechanisms of early adipocytic differentiation and may lead to development of therapeutic solutions for many general obesity issues.
Collapse
Affiliation(s)
- Yasuhiko Sera
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29401, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee DE, Kehlenbrink S, Lee H, Hawkins M, Yudkin JS. Getting the message across: mechanisms of physiological cross talk by adipose tissue. Am J Physiol Endocrinol Metab 2009; 296:E1210-29. [PMID: 19258492 DOI: 10.1152/ajpendo.00015.2009] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is associated with resistance of skeletal muscle to insulin-mediated glucose uptake, as well as resistance of different organs and tissues to other metabolic and vascular actions of insulin. In addition, the body is exquisitely sensitive to nutrient imbalance, with energy excess or a high-fat diet rapidly increasing insulin resistance, even before noticeable changes occur in fat mass. There is a growing acceptance of the fact that, as well as acting as a storage site for surplus energy, adipose tissue is an important source of signals relevant to, inter alia, energy homeostasis, fertility, and bone turnover. It has also been widely recognized that obesity is a state of low-grade inflammation, with adipose tissue generating substantial quantities of proinflammatory molecules. At a cellular level, the understanding of the signaling pathways responsible for such alterations has been intensively investigated. What is less clear, however, is how alterations of physiology, and of signaling, within one cell or one tissue are communicated to other parts of the body. The concepts of cell signals being disseminated systemically through a circulating "endocrine" signal have been complemented by the view that local signaling may similarly occur through autocrine or paracrine mechanisms. Yet, while much elegant work has focused on the alterations in signaling that are found in obesity or energy excess, there has been less attention paid to ways in which such signals may propagate to remote organs. This review of the integrative physiology of obesity critically appraises the data and outlines a series of hypotheses as to how interorgan cross talk takes place. The hypotheses presented include the "fatty acid hypothesis,", the "portal hypothesis,", the "endocrine hypothesis,", the "inflammatory hypothesis,", the "overflow hypothesis,", a novel "vasocrine hypothesis," and a "neural hypothesis," and the strengths and weaknesses of each hypothesis are discussed.
Collapse
Affiliation(s)
- Do-Eun Lee
- Department of Internal Medicine, Division of Endocrinology, Winthrop University Hospital, London, United Kingdom
| | | | | | | | | |
Collapse
|
44
|
Abstract
Specialized phagocytes are found in the most primitive multicellular organisms. Their roles in homeostasis and in distinguishing self from non-self have evolved with the complexity of organisms and their immune systems. Equally important, but often overlooked, are the roles of macrophages in tissue development. As discussed in this Review, these include functions in branching morphogenesis, neuronal patterning, angiogenesis, bone morphogenesis and the generation of adipose tissue. In each case, macrophage depletion impairs the formation of the tissue and compromises its function. I argue that in several diseases, the unrestrained acquisition of these developmental macrophage functions exacerbates pathology. For example, macrophages enhance tumour progression and metastasis by affecting tumour-cell migration and invasion, as well as angiogenesis.
Collapse
|
45
|
Lionetti L, Mollica MP, Lombardi A, Cavaliere G, Gifuni G, Barletta A. From chronic overnutrition to insulin resistance: the role of fat-storing capacity and inflammation. Nutr Metab Cardiovasc Dis 2009; 19:146-152. [PMID: 19171470 DOI: 10.1016/j.numecd.2008.10.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 10/09/2008] [Accepted: 10/31/2008] [Indexed: 12/17/2022]
Abstract
AIMS We analyze how the inflammatory state in adipose tissue caused by a condition of chronically positive energy balance can lead to insulin resistance first in adipose tissue, then in all insulin-sensitive tissues. DATA SYNTHESIS Chronic nutrient overload causes an increase in adipose depots that, if adipose tissue expandability is low, are characterized by an increased presence of hypertrophic adipocytes. This adipocyte hypertrophy is a possible stress condition for the endoplasmic reticulum (ER) that would lead to a proinflammatory state in adipose tissue. In this condition, ER stress would activate metabolic pathways that trigger insulin resistance, release of macrophage chemoattractant proteins, and in chronic inflammation, the death of the hypertrophic adipocyte. The infiltrated macrophages in turn release inflammatory proteins causing further recruitment of macrophages to adipose tissue and the release of inflammatory cytokines. Following these events, insulin resistance becomes extended to all adipose tissue. Insulin-resistant adipocytes, characterized by low liposynthetic capacity and high lipolytic capacity, cause increased release of free fatty acids (FFA). FFA released by lipolitic adipocytes may also activate Toll-like receptors 4 and then chemokines and cytokines release amplifying insulin resistance, lipolysis and inflammation in all adipose tissue. Moreover, increased circulating FFA levels, reduced circulating adiponectin levels and leptin resistance lead to decreased lipid oxidation in non-adipose tissues, thereby triggering ectopic accumulation of lipids, lipotoxicity and insulin resistance. CONCLUSION All the conditions that increase circulating fatty acids and cause lipid overloading (obesity, lipoatrophy, lipodystrophy, catabolic states, etc.) induce a lipotoxic state in non-adipose tissues that gives rise to insulin resistance.
Collapse
Affiliation(s)
- L Lionetti
- Department of Biological Sciences, Section of Physiology, University of Naples Federico II, Via Mezzocannone 8, 80134 Naples, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Macrophage-conditioned medium inhibits differentiation-induced Rb phosphorylation in 3T3-L1 preadipocytes. Exp Cell Res 2009; 315:411-8. [DOI: 10.1016/j.yexcr.2008.10.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 10/15/2008] [Accepted: 10/23/2008] [Indexed: 11/20/2022]
|
47
|
Structure of macrophage colony stimulating factor bound to FMS: diverse signaling assemblies of class III receptor tyrosine kinases. Proc Natl Acad Sci U S A 2008; 105:18267-72. [PMID: 19017797 DOI: 10.1073/pnas.0807762105] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Macrophage colony stimulating factor (M-CSF), through binding to its receptor FMS, a class III receptor tyrosine kinase (RTK), regulates the development and function of mononuclear phagocytes, and plays important roles in innate immunity, cancer and inflammation. We report a 2.4 A crystal structure of M-CSF bound to the first 3 domains (D1-D3) of FMS. The ligand binding mode of FMS is surprisingly different from KIT, another class III RTK, in which the major ligand-binding domain of FMS, D2, uses the CD and EF loops, but not the beta-sheet on the opposite side of the Ig domain as in KIT, to bind ligand. Calorimetric data indicate that M-CSF cannot dimerize FMS without receptor-receptor interactions mediated by FMS domains D4 and D5. Consistently, the structure contains only 1 FMS-D1-D3 molecule bound to a M-CSF dimer, due to a weak, hydrophilic M-CSF:FMS interface, and probably a conformational change of the M-CSF dimer in which binding to the second site is rendered unfavorable by FMS binding at the first site. The partial, intermediate complex suggests that FMS may be activated in two steps, with the initial engagement step distinct from the subsequent dimerization/activation step. Hence, the formation of signaling class III RTK complexes can be diverse, engaging various modes of ligand recognition and various mechanistic steps for dimerizing and activating receptors.
Collapse
|
48
|
Kishino T, Watanabe K, Urata T, Takano M, Uemura T, Nishikawa K, Mine Y, Matsumoto M, Ohtsuka K, Ohnishi H, Mori H, Takahashi S, Ishida H, Watanabe T. Visceral fat thickness in overweight men correlates with alterations in serum fatty acid composition. Clin Chim Acta 2008; 398:57-62. [PMID: 18771663 DOI: 10.1016/j.cca.2008.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2007] [Revised: 08/11/2008] [Accepted: 08/12/2008] [Indexed: 01/25/2023]
Abstract
BACKGROUND We examined relationships between visceral fat amount and alterations in serum fatty acid composition, both of which represent critical factors in the development of metabolic syndrome. METHODS Correlations were analyzed between visceral fat thickness as measured by ultrasonography and proportions of individual fatty acids in 21 normal-weight and 24 overweight Japanese men. RESULTS Significant associations were identified in overweight subjects. Visceral fat thickness displayed positive correlations to levels of palmitic acid and saturated fatty acids (r=0.475, P<0.05 and r=0.545, P<0.01, respectively); and negative correlations to levels of linoleic acid and polyunsaturated fatty acids (r=-0.513, P<0.05 and r=-0.428, P<0.05, respectively). Visceral fat thickness was also correlated with estimated desaturase activities, with positive correlations to Delta9- and Delta6-desaturase activities and negative correlations to Delta5-desaturase activity (r=0.580, P<0.01, r=0.669, P<0.01 and r=-0.559, P<0.01, respectively). No significant associations were identified in normal-weight subjects. CONCLUSIONS Significant associations between visceral fat amount and alterations in serum fatty acid composition were identified, but only in overweight individuals.
Collapse
Affiliation(s)
- Tomonori Kishino
- Department of Laboratory Medicine, Kyorin University School of Medicine, Tokyo 181-8611, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Although they were originally defined as haematopoietic-cell growth factors, colony-stimulating factors (CSFs) have been shown to have additional functions by acting directly on mature myeloid cells. Recent data from animal models indicate that the depletion of CSFs has therapeutic benefit in many inflammatory and/or autoimmune conditions and as a result, early-phase clinical trials targeting granulocyte/macrophage colony-stimulating factor and macrophage colony-stimulating factor have now commenced. The distinct biological features of CSFs offer opportunities for specific targeting, but with some associated risks. Here, I describe these biological features, discuss the probable specific outcomes of targeting CSFs in vivo and highlight outstanding questions that need to be addressed.
Collapse
|
50
|
Miller RS, Becker KG, Prabhu V, Cooke DW. Adipocyte gene expression is altered in formerly obese mice and as a function of diet composition. J Nutr 2008; 138:1033-8. [PMID: 18492830 PMCID: PMC2756972 DOI: 10.1093/jn/138.6.1033] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In the development of obesity, the source of excess energy may influence appetite and metabolism. To determine the effects of differences in diet composition in obesity, mice were fed either a high-carbohydrate diet (HC; 10% fat energy) or a high-fat energy-restricted diet (HFR; 60% fat energy) over 18 wk in weight-matched groups of mice. To identify obesity-associated genes with persistently altered expression following weight reduction, mice were fed either a standard low-fat diet (LF; 10% fat energy), an unrestricted high-fat diet (HF; 60% fat energy), or a HF diet followed by weight reduction (WR). Mice fed a HF diet had significantly greater gonadal fat mass and higher whole blood glucose concentrations than mice fed an HC diet. Of the mice fed a high-fat diet, total body weight and serum insulin concentrations were greater in HF than in HFR. Microarray analysis revealed that HF vs. HC feeding resulted in global differences in adipocyte gene expression patterns. Although we identified genes whose expression was altered in both moderately and severely obese mice, there were also a large number of genes with altered expression only in severe obesity. Formerly obese, WR mice did not differ significantly from lean controls in total body weight or physiological measures. However, microarray analysis revealed distinctly different patterns of adipocyte gene expression. Furthermore, there were 398 genes with altered expression in HF mice that persisted in WR mice. Genes with persistently altered expression following obesity may play a role in rebound weight gain following weight reduction.
Collapse
Affiliation(s)
- Ryan S. Miller
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, Ilyssa Center for Molecular and Cellular Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD 21287,To whom correspondence should be addressed. E-mail:
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, National Institute on Aging, NIH, Baltimore, MD 21224
| | - Vinayakumar Prabhu
- Gene Expression and Genomics Unit, National Institute on Aging, NIH, Baltimore, MD 21224
| | - David W. Cooke
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, Ilyssa Center for Molecular and Cellular Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|