1
|
You Z, Hu Z, Hou C, Ma C, Xu X, Zheng Y, Sun X, Ke Y, Liang J, Xie Z, Shu L, Liu Y. FABP4 facilitates epithelial-mesenchymal transition via elevating CD36 expression in glioma cells. Neoplasia 2024; 57:101050. [PMID: 39243502 PMCID: PMC11406018 DOI: 10.1016/j.neo.2024.101050] [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: 05/26/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis. A better understanding of mechanisms concerned in glioma invasion might be critical for treatment optimization. Given that epithelial-mesenchymal transition in tumor cells is closely associated with glioma progression and recurrence, identifying pivotal mediators in GBM EMT process is urgently needed. As a member of Fatty acid binding protein (FABP) family, FABP4 serves as chaperones for free fatty acids and participates in cellular process including fatty acid uptake, transport, and metabolism. In this study, our data revealed that FABP4 expression was elevated in human GBM samples and correlated with a mesenchymal glioma subtype. Gain of function and loss of function experiments indicated that FABP4 potently rendered glioma cells increased filopodia formation and cell invasiveness. Differential expression genes analysis and GSEA in TCGA dataset revealed an EMT-related molecular signature in FABP4-mediated signaling pathways. Cell interaction analysis suggested CD36 as a potential target regulated by FABP4. Furthermore, in vitro mechanistic experiments demonstrated that FABP4-induced CD36 expression promoted EMT via non-canonical TGFβ pathways. An intracranial glioma model was constructed to assess the effect of FABP4 on tumor progression in vivo. Together, our findings demonstrated a critical role for FABP4 in the regulation invasion and EMT in GBM, and suggest that pharmacological inhibition of FABP4 may represent a promising therapeutic strategy for treatment of GBM.
Collapse
Affiliation(s)
- Zhongsheng You
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zihao Hu
- School of Medicine, Nankai University, Tianjin, PR China
| | - Chongxian Hou
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Chengcheng Ma
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China; School of Medicine, Nankai University, Tianjin, PR China
| | - Xiangdong Xu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yaofeng Zheng
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Xinlin Sun
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yiquan Ke
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Jianli Liang
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zijing Xie
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China.
| | - Yang Liu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China.
| |
Collapse
|
2
|
Zhang D, Wang M, Liu G, Li X, Yu W, Hui Z, Ren X, Sun Q. Novel FABP4 +C1q + macrophages enhance antitumor immunity and associated with response to neoadjuvant pembrolizumab and chemotherapy in NSCLC via AMPK/JAK/STAT axis. Cell Death Dis 2024; 15:717. [PMID: 39353883 PMCID: PMC11445384 DOI: 10.1038/s41419-024-07074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024]
Abstract
Immune checkpoint inhibitors (ICIs) immunotherapy facilitates new approaches to achieve precision cancer treatment. A growing number of patients with non-small cell lung cancer (NSCLC) have benefited from treatment with neoadjuvant ICIs combined with chemotherapy. However, the mechanisms and associations between the therapeutic efficacy of neoadjuvant pembrolizumab and chemotherapy (NAPC) and macrophage subsets are still unclear. We performed single-cell RNA sequencing (scRNA-seq) and identified a novel FABP4+C1q+ macrophage subtype, which exhibited stronger proinflammatory cytokine production and phagocytic ability. This subtype was found to be more abundant in tumor tissues and lymph nodes of major pathological response (MPR) patients compared to non-MPR patients, and was associated with a good efficacy of NAPC. Multiplex fluorescent immunohistochemical (mIHC) staining was subsequently used to verify our findings. Further mechanistic studies indicated that FABP4 and C1q regulate the expression of proinflammatory cytokines synergistically. In addition, FABP4 and C1q promote fatty acid synthesis, enhance anti-apoptosis ability and phagocytic ability of macrophage via the interaction of AMPK/JAK/STAT axis. This study provides novel insights into the underlying mechanisms and predictive biomarkers of NAPC. Our findings contribute to improving the prognosis of patients with NSCLC by potentially guiding more precise patient selection and treatment strategies. NOVELTY & IMPACT STATEMENTS: We identified a group of macrophages (FABP4+C1q+ macrophages) related to the therapeutic efficacy of neoadjuvant chemoimmunotherapy. FABP4+C1q+ macrophages highly expressed proinflammatory cytokines-related genes and had a strong cytokine production and phagocytic ability. We believe that our study provides a novel insight into the synergistic mechanism of neoadjuvant ICI combined with chemotherapy and may lead to improved clinical outcomes in patients with NSCLC in the future.
Collapse
Affiliation(s)
- Dong Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Min Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gen Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xin Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenzhen Hui
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| | - Qian Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
3
|
Warren WG, Osborn M, Duffy P, Yates A, O'Sullivan SE. Potential safety implications of fatty acid-binding protein inhibition. Toxicol Appl Pharmacol 2024; 491:117079. [PMID: 39218163 DOI: 10.1016/j.taap.2024.117079] [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: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Fatty acid-binding proteins (FABPs) are small intracellular proteins that regulate fatty acid metabolism, transport, and signalling. There are ten known human isoforms, many of which are upregulated and involved in clinical pathologies. As such, FABP inhibition may be beneficial in disease states such as cancer, and those involving the cardiovascular system, metabolism, immunity, and cognition. Recently, a potent, selective FABP5 inhibitor (ART26.12), with 90-fold selectivity to FABP3 and 20-fold selectivity to FABP7, was found to be remarkably benign, with a no-observed-adverse-effect level of 1000 mg/kg in rats and dogs, showing no genotoxicity, cardiovascular, central, or respiratory toxicity. To understand the potential implication of FABP inhibition more fully, this review systematically assessed literature investigating genetic knockout, knockdown, and pharmacological inhibition of FABP3, FABP4, FABP5, or FABP7. Analysis of the literature revealed that animals bred not to express FABPs showed the most biological effects, suggesting key roles of these proteins during development. FABP ablation sometimes exacerbated symptoms of disease models, particularly those linked to metabolism, inflammatory and immune responses, cardiac contractility, neurogenesis, and cognition. However, FABP inhibition (genetic silencing or pharmacological) had a positive effect in many more disease conditions. Several polymorphisms of each FABP gene have also been linked to pathological conditions, but it was unclear how several polymorphisms affected protein function. Overall, analysis of the literature to date suggests that pharmacological inhibition of FABPs in adults is of low risk.
Collapse
Affiliation(s)
- William G Warren
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom.
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Paul Duffy
- Apconix Ltd., Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | | |
Collapse
|
4
|
Wu X, Cheong LY, Yuan L, Jin L, Zhang Z, Xiao Y, Zhou Z, Xu A, Hoo RLC, Shu L. Islet-Resident Memory T Cells Orchestrate the Immunopathogenesis of Type 1 Diabetes through the FABP4-CXCL10 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308461. [PMID: 38884133 PMCID: PMC11321687 DOI: 10.1002/advs.202308461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/18/2024] [Indexed: 06/18/2024]
Abstract
Type 1 diabetes (T1D) is a chronic disease characterized by self-destruction of insulin-producing pancreatic β cells by cytotoxic T cell activity. However, the pathogenic mechanism of T cell infiltration remains obscure. Recently, tissue-resident memory T (TRM) cells have been shown to contribute to cytotoxic T cell recruitment. TRM cells are found present in human pancreas and are suggested to modulate immune homeostasis. Here, the role of TRM cells in the development of T1D is investigated. The presence of TRM cells in pancreatic islets is observed in non-obese diabetic (NOD) mice before T1D onset. Mechanistically, elevated fatty acid-binding protein 4 (FABP4) potentiates the survival and alarming function of TRM cells by promoting fatty acid utilization and C-X-C motif chemokine 10 (CXCL10) secretion, respectively. In NOD mice, genetic deletion of FABP4 or depletion of TRM cells using CD69 neutralizing antibodies resulted in a similar reduction of pancreatic cytotoxic T cell recruitment, a delay in diabetic incidence, and a suppression of CXCL10 production. Thus, targeting FABP4 may represent a promising therapeutic strategy for T1D.
Collapse
Affiliation(s)
- Xiaoping Wu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of Pharmacology & PharmacyThe University of Hong KongHong Kong999077P. R. China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of MedicineThe University of Hong KongHong Kong999077P. R. China
| | - Lufengzi Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of Pharmacology & PharmacyThe University of Hong KongHong Kong999077P. R. China
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of MedicineThe University of Hong KongHong Kong999077P. R. China
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of Pharmacology & PharmacyThe University of Hong KongHong Kong999077P. R. China
| | - Yang Xiao
- Second Xiangya HospitalKey Laboratory of Diabetes ImmunologyNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunan410083P. R. China
| | - Zhiguang Zhou
- Second Xiangya HospitalKey Laboratory of Diabetes ImmunologyNational Clinical Research Center for Metabolic DiseasesCentral South UniversityChangshaHunan410083P. R. China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of Pharmacology & PharmacyThe University of Hong KongHong Kong999077P. R. China
- Department of MedicineThe University of Hong KongHong Kong999077P. R. China
| | - Ruby LC Hoo
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- Department of Pharmacology & PharmacyThe University of Hong KongHong Kong999077P. R. China
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077P. R. China
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for Cancer, Department of Hematological OncologySun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
5
|
Schonblum A, Ali Naser D, Ovadia S, Egbaria M, Puyesky S, Epshtein A, Wald T, Mercado-Medrez S, Ashery-Padan R, Landsman L. Beneficial islet inflammation in health depends on pericytic TLR/MyD88 signaling. J Clin Invest 2024; 134:e179335. [PMID: 38885342 PMCID: PMC11245159 DOI: 10.1172/jci179335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects β cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and β cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1β production. This aberrant pericyte-orchestrated islet inflammation was associated with β cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1β production. Treatment with either Cxcl1 or IL-1β restored the mature β cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Anat Schonblum
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Dunia Ali Naser
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Shai Ovadia
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Mohammed Egbaria
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Shani Puyesky
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Alona Epshtein
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Tomer Wald
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Sophia Mercado-Medrez
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| |
Collapse
|
6
|
Yang M, Shu W, Zhai X, Yang X, Zhou H, Pan B, Li C, Lu D, Cai J, Zheng S, Jin B, Wei X, Xu X. Integrated multi-omic analysis identifies fatty acid binding protein 4 as a biomarker and therapeutic target of ischemia-reperfusion injury in steatotic liver transplantation. Cell Mol Life Sci 2024; 81:83. [PMID: 38341383 PMCID: PMC10858962 DOI: 10.1007/s00018-023-05110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND AND AIMS Due to a lack of donor grafts, steatotic livers are used more often for liver transplantation (LT). However, steatotic donor livers are more sensitive to ischemia-reperfusion (IR) injury and have a worse prognosis after LT. Efforts to optimize steatotic liver grafts by identifying injury targets and interventions have become a hot issue. METHODS Mouse LT models were established, and 4D label-free proteome sequencing was performed for four groups: normal control (NC) SHAM, high-fat (HF) SHAM, NC LT, and HF LT to screen molecular targets for aggravating liver injury in steatotic LT. Expression detection of molecular targets was performed based on liver specimens from 110 donors to verify its impact on the overall survival of recipients. Pharmacological intervention using small-molecule inhibitors on an injury-related target was used to evaluate the therapeutic effect. Transcriptomics and metabolomics were performed to explore the regulatory network and further integrated bioinformatics analysis and multiplex immunofluorescence were adopted to assess the regulation of pathways and organelles. RESULTS HF LT group represented worse liver function compared with NC LT group, including more apoptotic hepatocytes (P < 0.01) and higher serum transaminase (P < 0.05). Proteomic results revealed that the mitochondrial membrane, endocytosis, and oxidative phosphorylation pathways were upregulated in HF LT group. Fatty acid binding protein 4 (FABP4) was identified as a hypoxia-inducible protein (fold change > 2 and P < 0.05) that sensitized mice to IR injury in steatotic LT. The overall survival of recipients using liver grafts with high expression of FABP4 was significantly worse than low expression of FABP4 (68.5 vs. 87.3%, P < 0.05). Adoption of FABP4 inhibitor could protect the steatotic liver from IR injury during transplantation, including reducing hepatocyte apoptosis, reducing serum transaminase (P < 0.05), and alleviating oxidative stress damage (P < 0.01). According to integrated transcriptomics and metabolomics analysis, cAMP signaling pathway was enriched following FABP4 inhibitor use. The activation of cAMP signaling pathway was validated. Microscopy and immunofluorescence staining results suggested that FABP4 inhibitors could regulate mitochondrial membrane homeostasis in steatotic LT. CONCLUSIONS FABP4 was identified as a hypoxia-inducible protein that sensitized steatotic liver grafts to IR injury. The FABP4 inhibitor, BMS-309403, could activate of cAMP signaling pathway thereby modulating mitochondrial membrane homeostasis, reducing oxidative stress injury in steatotic donors.
Collapse
Affiliation(s)
- Mengfan Yang
- Department of Organ Transplantation, Qilu Hospital of Shandong University, Jinan, 250012, China
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Wenzhi Shu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital, Shandong University, Jinan, 250033, China
| | - Xinyu Yang
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Huaxin Zhou
- Department of Hepatobiliary Surgery, The Second Hospital, Shandong University, Jinan, 250033, China
| | - Binhua Pan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Changbiao Li
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Di Lu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jinzhen Cai
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, 266035, China
| | - Shusen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Bin Jin
- Department of Organ Transplantation, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Department of Hepatobiliary Surgery, The Second Hospital, Shandong University, Jinan, 250033, China.
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310003, China.
| |
Collapse
|
7
|
Li Y, Tang C, Vanarsa K, Thai N, Castillo J, Lea GAB, Lee KH, Kim S, Pedroza C, Wu T, Saxena R, Mok CC, Mohan C. Proximity extension assay proteomics and renal single cell transcriptomics uncover novel urinary biomarkers for active lupus nephritis. J Autoimmun 2024; 143:103165. [PMID: 38194790 DOI: 10.1016/j.jaut.2023.103165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024]
Abstract
OBJECTIVE To identify urinary biomarkers that can distinguish active renal involvement in Lupus Nephritis (LN), a severe manifestation of systemic lupus erythematosus (SLE). METHODS Urine from 117 subjects, comprised of inactive SLE, active non-renal lupus, active LN, and healthy controls, were subjected to Proximity Extension Assay (PEA) based comprehensive proteomics followed by ELISA validation in an independent, ethnically diverse cohort. Proteomic data is also cross-referenced to renal transcriptomic data to elucidate cellular origins of biomarkers. RESULTS Systems biology analyses revealed progressive activation of cytokine signaling, chemokine activity and coagulation pathways, with worsening renal disease. In addition to validating 30 previously reported biomarkers, this study uncovers several novel candidates. Following ELISA validation in an independent cohort of different ethnicity, the six most discriminatory biomarkers for active LN were urinary ICAM-2, FABP4, FASLG, IGFBP-2, SELE and TNFSF13B/BAFF, with ROC AUC ≥80%, with most correlating strongly with clinical disease activity. Transcriptomic analyses of LN kidneys mapped the likely origin of these proteins to intra-renal myeloid cells (CXCL16, IL-1RT2, TNFSF13B/BAFF), T/NK cells (FASLG), leukocytes (ICAM2) and endothelial cells (SELE). CONCLUSION In addition to confirming the diagnostic potential of urine ALCAM, CD163, MCP1, SELL, ICAM1, VCAM1, NGAL and TWEAK for active LN, this study adds urine ICAM-2, FABP4, FASLG, IGFBP-2, SELE, and TNFSF13B/BAFF as additional markers that warrant systematic validation in larger cross-sectional and longitudinal cohorts.
Collapse
Affiliation(s)
- Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Nga Thai
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Jessica Castillo
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | | | - Kyung Hyun Lee
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Soojin Kim
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Claudia Pedroza
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Ramesh Saxena
- Department of Internal Medicine, Division of Nephrology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, New Territories, Hong Kong, China
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
8
|
Ubba V, Joseph S, Awe O, Jones D, Dsilva MK, Feng M, Wang J, Fu X, Akbar RJ, Bodnar BH, Hu W, Wang H, Yang X, Yang L, Yang P, Taib B, Ahima R, Divall S, Wu S. Reproductive Profile of Neuronal Androgen Receptor Knockout Female Mice With a Low Dose of DHT. Endocrinology 2024; 165:bqad199. [PMID: 38156784 PMCID: PMC10794876 DOI: 10.1210/endocr/bqad199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/08/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Hyperandrogenism and polycystic ovarian syndrome result from the imbalance or increase of androgen levels in females. Androgen receptor (AR) mediates the effects of androgens, and this study examines whether neuronal AR plays a role in reproduction under normal and increased androgen conditions in female mice. The neuron-specific AR knockout (KO) mouse (SynARKO) was generated from a female mouse (synapsin promoter driven Cre) and a male mouse (Ar fl/y). Puberty onset and the levels of reproductive hormones such as LH, FSH, testosterone, and estradiol were comparable between the control and the SynARKO mice. There were no differences in cyclicity and fertility between the control and SynARKO mice, with similar impairment in both groups on DHT treatment. Neuronal AR KO, as in this SynARKO mouse model, did not alleviate the infertility associated with DHT treatment. These studies suggest that neuronal AR KO neither altered reproductive function under physiological androgen levels, nor restored fertility under hyperandrogenic conditions.
Collapse
Affiliation(s)
- Vaibhave Ubba
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Serene Joseph
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Dustin Jones
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Milan K Dsilva
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Mingxiao Feng
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
| | - Junjiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
- Departments of Gastrointestinal Surgery and General Surgery, Guangdong Provincial People’s Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Xiaomin Fu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Razeen J Akbar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Brittany H Bodnar
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Wenhui Hu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Ling Yang
- Department of Medical Genetics & Molecular Biochemistry, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, 21201, USA
| | - Bouchra Taib
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Rexford Ahima
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle’s Children’s Hospital, Seattle, WA, 98145-5005, USA
| | - Sheng Wu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21087, USA
| |
Collapse
|
9
|
Xiong Z, Xu X, Zhang Y, Ma C, Hou C, You Z, Shu L, Ke Y, Liu Y. IFITM3 promotes glioblastoma stem cell-mediated angiogenesis via regulating JAK/STAT3/bFGF signaling pathway. Cell Death Dis 2024; 15:45. [PMID: 38218875 PMCID: PMC10787840 DOI: 10.1038/s41419-023-06416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/15/2024]
Abstract
Interferon-induced transmembrane protein 3 (IFITM3) has been previously verified to be an endosomal protein that prevents viral infection. Recent findings suggested IFITM3 as a key factor in tumor invasion and progression. To clarify the role and molecular mechanism of IFITM3 in Glioblastoma multiforme (GBM) progression, we investigated the expression of IFITM3 in glioma datasets culled from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA). Primary GBM stem cells (GSCs) were cultured and identified in vitro. Loss-of-function and gain-of-function experiments were established by using shRNAs and lentiviral vectors targeting IFITM3. Co-culture system of GSCs and vascular endothelial cells was constructed in a Transwell chamber. Tube formation and spheroid-based angiogenesis assays were performed to determine the angiogenic capacity of endothelial cells. Results revealed that IFITM3 is elevated in GBM samples and predictive of adverse outcome. Mechanistically, GSCs-derived IFITM3 causes activation of Jak2/STAT3 signaling and leads to robust secretion of bFGF into tumor environment, which eventually results in enhanced angiogenesis. Taken together, these evidence indicated IFITM3 as an essential factor in GBM angiogenesis. Our findings provide a new insight into mechanism by which IFITM3 modulates GBM angiogenesis.
Collapse
Affiliation(s)
- Zhangsheng Xiong
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Xiangdong Xu
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Yuxuan Zhang
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Chengcheng Ma
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Chongxian Hou
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Zhongsheng You
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
- Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Guangzhou, PR China.
| | - Yiquan Ke
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China.
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China.
| | - Yang Liu
- Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510060, PR China.
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou, 510060, PR China.
| |
Collapse
|
10
|
Huang Y, Cui X, Li Z, Yuan S, Han Y, Xu X, Fu X, Shi K, Zhang Z, Wei J, Xia S, Xiao Y, Xue S, Sun L, Liu H, Zhu X. High-expression of FABP4 in Tubules is a Risk Factor for Poor Prognosis in DKD Patients. Curr Med Chem 2024; 31:3436-3446. [PMID: 38299395 DOI: 10.2174/0109298673268265231228125431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Lipid metabolism imbalance is involved in the mechanism of renal tubular injury in diabetic kidney disease (DKD). Fatty acid binding protein 4 (FABP4) has been reported to participate in cellular lipid toxicity. However, the expression of FABP4 in renal tissues of DKD and its correlation with clinical/ pathological parameters and prognosis have not been studied. METHODS A retrospective cohort study was conducted in 108 hospitalized Type 2 diabetes (T2D) patients with renal injury, including 70 with DKD and 38 with NDKD (non-DKD). Clinical features, pathological findings, and follow-up parameters were collected. Serum and urine FABP4 were detected by ELISA. An immunohistochemistry stain was used to determine FABP4 in renal tubulointerstitium. A double immunofluorescence stain was employed to assess FABP4- and CD68-positive macrophages. Correlation analysis, logistic regression models, receiver operating characteristic (ROC), and Kaplan-Meier survival curve were performed for statistical analysis. RESULTS DKD patients had increased expression of FABP4 and ectopic fat deposition in tubules. As shown by correlation analyses, FABP4 expression in renal tubules was positively correlated with UNAG (r=0.589, p=0.044) and ESRD (r=0.740, p=0.004). Multivariate regression analysis revealed that UNAG level was correlated with FABP4 expression level above median value (odds ratio:1.154, 95% confidence interval:1.009-1.321, p=0.037). High-expression of FABP4 in renal tubules of DKD was at an increased risk of ESRD. Increased FABP4 expression in inflammatory cells was also associated with ESRD in DKD. CONCLUSION High-expression of FABP4 is involved in the pathogenesis of renal tubular lipid injury and is a risk factor for poor prognosis in DKD patients.
Collapse
Affiliation(s)
- Yao Huang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xinyuan Cui
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zheng Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shuguang Yuan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xiangqing Xu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xiao Fu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Kewen Shi
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zurong Zhang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Jinying Wei
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shiyu Xia
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yang Xiao
- Key Laboratory of Diabetes Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Song Xue
- Key Laboratory of Diabetes Immunology, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, China
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xuejing Zhu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
11
|
Shi Y, Wang CC, Wu L, Zhang Y, Xu A, Wang Y. Pathophysiological Insight into Fatty Acid-Binding Protein-4: Multifaced Roles in Reproduction, Pregnancy, and Offspring Health. Int J Mol Sci 2023; 24:12655. [PMID: 37628833 PMCID: PMC10454382 DOI: 10.3390/ijms241612655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Fatty acid-binding protein-4 (FABP4), commonly known as adipocyte-fatty acid-binding protein (A-FABP), is a pleiotropic adipokine that broadly affects immunity and metabolism. It has been increasingly recognized that FABP4 dysfunction is associated with various metabolic syndromes, including obesity, diabetes, cardiovascular diseases, and metabolic inflammation. However, its explicit roles within the context of women's reproduction and pregnancy remain to be investigated. In this review, we collate recent studies probing the influence of FABP4 on female reproduction, pregnancy, and even fetal health. Elevated circulating FABP4 levels have been found to correlate with impaired reproductive function in women, such as polycystic ovary syndrome and endometriosis. Throughout pregnancy, FABP4 affects maternal-fetal interface homeostasis by affecting both glycolipid metabolism and immune tolerance, leading to adverse pregnancy outcomes, including miscarriage, gestational obesity, gestational diabetes, and preeclampsia. Moreover, maternal FABP4 levels exhibit a substantial linkage with the metabolic health of offspring. Herein, we discuss the emerging significance and potential application of FABP4 in reproduction and pregnancy health and delve into its underlying mechanism at molecular levels.
Collapse
Affiliation(s)
- Yue Shi
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Chi-Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
- Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Liqun Wu
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China;
| | - Yunqing Zhang
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong;
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
| |
Collapse
|
12
|
Prentice KJ, Lee A, Cedillo P, Inouye KE, Ertunc ME, Riveros JK, Lee GY, Hotamisligil GS. Sympathetic tone dictates the impact of lipolysis on FABP4 secretion. J Lipid Res 2023; 64:100386. [PMID: 37172691 PMCID: PMC10248869 DOI: 10.1016/j.jlr.2023.100386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Levels of circulating fatty acid binding protein 4 (FABP4) protein are strongly associated with obesity and metabolic disease in both mice and humans, and secretion is stimulated by β-adrenergic stimulation both in vivo and in vitro. Previously, lipolysis-induced FABP4 secretion was found to be significantly reduced upon pharmacological inhibition of adipose triglyceride lipase (ATGL) and was absent from adipose tissue explants from mice specifically lacking ATGL in their adipocytes (ATGLAdpKO). Here, we find that upon activation of β-adrenergic receptors in vivo, ATGLAdpKO mice unexpectedly exhibited significantly higher levels of circulating FABP4 as compared with ATGLfl/fl controls, despite no corresponding induction of lipolysis. We generated an additional model with adipocyte-specific deletion of both FABP4 and ATGL (ATGL/FABP4AdpKO) to evaluate the cellular source of this circulating FABP4. In these animals, there was no evidence of lipolysis-induced FABP4 secretion, indicating that the source of elevated FABP4 levels in ATGLAdpKO mice was indeed from the adipocytes. ATGLAdpKO mice exhibited significantly elevated corticosterone levels, which positively correlated with plasma FABP4 levels. Pharmacological inhibition of sympathetic signaling during lipolysis using hexamethonium or housing mice at thermoneutrality to chronically reduce sympathetic tone significantly reduced FABP4 secretion in ATGLAdpKO mice compared with controls. Therefore, activity of a key enzymatic step of lipolysis mediated by ATGL, per se, is not required for in vivo stimulation of FABP4 secretion from adipocytes, which can be induced through sympathetic signaling.
Collapse
Affiliation(s)
- Kacey J Prentice
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexandra Lee
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Paulina Cedillo
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Karen E Inouye
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Meric Erikci Ertunc
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jillian K Riveros
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Grace Yankun Lee
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gökhan S Hotamisligil
- Department of Molecular Metabolism; Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
13
|
Burganova G, Schonblum A, Sakhneny L, Epshtein A, Wald T, Tzaig M, Landsman L. Pericytes modulate islet immune cells and insulin secretion through Interleukin-33 production in mice. Front Endocrinol (Lausanne) 2023; 14:1142988. [PMID: 36967785 PMCID: PMC10034381 DOI: 10.3389/fendo.2023.1142988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction Immune cells were recently shown to support β-cells and insulin secretion. However, little is known about how islet immune cells are regulated to maintain glucose homeostasis. Administration of various cytokines, including Interleukin-33 (IL-33), was shown to influence β-cell function. However, the role of endogenous, locally produced IL-33 in pancreatic function remains unknown. Here, we show that IL-33, produced by pancreatic pericytes, is required for glucose homeostasis. Methods To characterize pancreatic IL-33 production, we employed gene expression, flow cytometry, and immunofluorescence analyses. To define the role of this cytokine, we employed transgenic mouse systems to delete the Il33 gene selectively in pancreatic pericytes, in combination with the administration of recombinant IL-33. Glucose response was measured in vivo and in vitro, and morphometric and molecular analyses were used to measure β-cell mass and gene expression. Immune cells were analyzed by flow cytometry. Resuts Our results show that pericytes are the primary source of IL-33 in the pancreas. Mice lacking pericytic IL-33 were glucose intolerant due to impaired insulin secretion. Selective loss of pericytic IL-33 was further associated with reduced T and dendritic cell numbers in the islets and lower retinoic acid production by islet macrophages. Discussion Our study demonstrates the importance of local, pericytic IL-33 production for glucose regulation. Additionally, it proposes that pericytes regulate islet immune cells to support β-cell function in an IL-33-dependent manner. Our study reveals an intricate cellular network within the islet niche.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
14
|
Exploring the Anti-Inflammatory Effect of Inulin by Integrating Transcriptomic and Proteomic Analyses in a Murine Macrophage Cell Model. Nutrients 2023; 15:nu15040859. [PMID: 36839217 PMCID: PMC9965215 DOI: 10.3390/nu15040859] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Inulin is a natural polysaccharide classified as a soluble fiber with demonstrated prebiotic activity. Prebiotics can reduce intestinal and systemic inflammation through modulation of the gut microflora and their metabolites. Additionally, extensive research is illuminating the role of macrophages in the interaction between gut microbiota and many systemic inflammatory diseases. In this study, the anti-inflammatory properties of inulin were evaluated using a murine macrophage cell model (RAW 264.7) of inflammation, and the immunomodulatory mechanism was investigated using omics technologies. The cells underwent comprehensive transcriptomic and proteomic analyses to identify the mechanisms responsible for the observed anti-inflammatory phenotype. Functional analyses of these omics results revealed two potential mechanisms that may lead to an overall reduction in cytokine and chemokine transcription: the inhibition of the NF-κB signaling pathway, leading to the downregulation of proinflammatory factors such as COX2, and the promotion of the phase II defense protein Hmox1 via the Nrf2 pathway. This study provides promising targets for research on immune modulation by dietary fibers and offers new strategies for the design of functional ingredients, foods, and nutraceutical products, which could ultimately lead to personalized nutrition and improved consumer health.
Collapse
|
15
|
Chekol Abebe E, Tilahun Muche Z, Behaile T/Mariam A, Mengie Ayele T, Mekonnen Agidew M, Teshome Azezew M, Abebe Zewde E, Asmamaw Dejenie T, Asmamaw Mengstie M. The structure, biosynthesis, and biological roles of fetuin-A: A review. Front Cell Dev Biol 2022; 10:945287. [PMID: 35923855 PMCID: PMC9340150 DOI: 10.3389/fcell.2022.945287] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 11/21/2022] Open
Abstract
Fetuin-A is a heterodimeric plasma glycoprotein containing an A-chain of 282 amino acids and a B-chain of 27 amino acid residues linked by a single inter-disulfide bond. It is predominantly expressed in embryonic cells and adult hepatocytes, and to a lesser extent in adipocytes and monocytes. Fetuin-A binds with a plethora of receptors and exhibits multifaceted physiological and pathological functions. It is involved in the regulation of calcium metabolism, osteogenesis, and the insulin signaling pathway. It also acts as an ectopic calcification inhibitor, protease inhibitor, inflammatory mediator, anti-inflammatory partner, atherogenic factor, and adipogenic factor, among other several moonlighting functions. Fetuin-A has also been demonstrated to play a crucial role in the pathogenesis of several disorders. This review mainly focuses on the structure, synthesis, and biological roles of fetuin-A. Information was gathered manually from various journals via electronic searches using PubMed, Google Scholar, HINARI, and Cochrane Library from inception to 2022. Studies written in English and cohort, case-control, cross-sectional, or experimental studies were considered in the review, otherwise excluded.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Zelalem Tilahun Muche
- Department of Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Awigchew Behaile T/Mariam
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Teklie Mengie Ayele
- Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Melaku Mekonnen Agidew
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Muluken Teshome Azezew
- Department of Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Edgeit Abebe Zewde
- Department of Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Misganaw Asmamaw Mengstie
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
16
|
Interplay between Fatty Acid Binding Protein 4, Fetuin-A, Retinol Binding Protein 4 and Thyroid Function in Metabolic Dysregulation. Metabolites 2022; 12:metabo12040300. [PMID: 35448487 PMCID: PMC9026429 DOI: 10.3390/metabo12040300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 12/22/2022] Open
Abstract
Signalling between the tissues integrating synthesis, transformation and utilization of energy substrates and their regulatory hormonal axes play a substantial role in the development of metabolic disorders. Interactions between cytokines, particularly liver derived hepatokines and adipokines, secreted from adipose tissue, constitute one of major areas of current research devoted to metabolic dysregulation. The thyroid exerts crucial influence on the maintenance of basal metabolic rate, thermogenesis, carbohydrate and lipid metabolism, while its dysfunction promotes the development of metabolic disorders. In this review, we discuss the interplay between three adipokines: fatty acid binding protein type 4, fetuin-A, retinol binding protein type 4 and thyroid hormones, that shed a new light onto mechanisms underlying atherosclerosis, cardiovascular complications, obesity, insulin resistance and diabetes accompanying thyroid dysfunction. Furthermore, we summarize clinical findings on those cytokines in the course of thyroid disorders.
Collapse
|
17
|
Prentice KJ, Saksi J, Robertson LT, Lee GY, Inouye KE, Eguchi K, Lee A, Cakici O, Otterbeck E, Cedillo P, Achenbach P, Ziegler AG, Calay ES, Engin F, Hotamisligil GS. A hormone complex of FABP4 and nucleoside kinases regulates islet function. Nature 2021; 600:720-726. [PMID: 34880500 DOI: 10.1038/s41586-021-04137-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/14/2021] [Indexed: 11/09/2022]
Abstract
The liberation of energy stores from adipocytes is critical to support survival in times of energy deficit; however, uncontrolled or chronic lipolysis associated with insulin resistance and/or insulin insufficiency disrupts metabolic homeostasis1,2. Coupled to lipolysis is the release of a recently identified hormone, fatty-acid-binding protein 4 (FABP4)3. Although circulating FABP4 levels have been strongly associated with cardiometabolic diseases in both preclinical models and humans4-7, no mechanism of action has yet been described8-10. Here we show that hormonal FABP4 forms a functional hormone complex with adenosine kinase (ADK) and nucleoside diphosphate kinase (NDPK) to regulate extracellular ATP and ADP levels. We identify a substantial effect of this hormone on beta cells and given the central role of beta-cell function in both the control of lipolysis and development of diabetes, postulate that hormonal FABP4 is a key regulator of an adipose-beta-cell endocrine axis. Antibody-mediated targeting of this hormone complex improves metabolic outcomes, enhances beta-cell function and preserves beta-cell integrity to prevent both type 1 and type 2 diabetes. Thus, the FABP4-ADK-NDPK complex, Fabkin, represents a previously unknown hormone and mechanism of action that integrates energy status with the function of metabolic organs, and represents a promising target against metabolic disease.
Collapse
Affiliation(s)
- Kacey J Prentice
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Jani Saksi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Lauren T Robertson
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Grace Y Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Karen E Inouye
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Kosei Eguchi
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Alexandra Lee
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Ozgur Cakici
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Emily Otterbeck
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Paulina Cedillo
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Ediz S Calay
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Feyza Engin
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA.,Departments of Biomolecular Chemistry and Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Harvard T. H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
18
|
Yang Z, Yang D, Tan F, Wong CW, Yang JY, Zhou D, Cai Z, Lin SH. Multi-Omics Comparison of the Spontaneous Diabetes Mellitus and Diet-Induced Prediabetic Macaque Models. Front Pharmacol 2021; 12:784231. [PMID: 34880765 PMCID: PMC8645867 DOI: 10.3389/fphar.2021.784231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
The prevalence of diabetes mellitus has been increasing for decades worldwide. To develop safe and potent therapeutics, animal models contribute a lot to the studies of the mechanisms underlying its pathogenesis. Dietary induction using is a well-accepted protocol in generating insulin resistance and diabetes models. In the present study, we reported the multi-omics profiling of the liver and sera from both peripheral blood and hepatic portal vein blood from Macaca fascicularis that spontaneously developed Type-2 diabetes mellitus with a chow diet (sDM). The other two groups of the monkeys fed with chow diet and high-fat high-sugar (HFHS) diet, respectively, were included for comparison. Analyses of various omics datasets revealed the alterations of high consistency. Between the sDM and HFHS monkeys, both the similar and unique alterations in the lipid metabolism have been demonstrated from metabolomic, transcriptomic, and proteomic data repeatedly. The comparison of the proteome and transcriptome confirmed the involvement of fatty acid binding protein 4 (FABP4) in the diet-induced pathogenesis of diabetes in macaques. Furthermore, the commonly changed genes between spontaneous diabetes and HFHS diet-induced prediabetes suggested that the alterations in the intra- and extracellular structural proteins and cell migration in the liver might mediate the HFHS diet induction of diabetes mellitus.
Collapse
Affiliation(s)
- Zhu Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Dianqiang Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Fancheng Tan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Chi Wai Wong
- Guangzhou Huazhen Biosciences Co., Ltd., Guangzhou, China
| | - James Y. Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Xiamen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
19
|
Dahlström EH, Saksi J, Forsblom C, Uglebjerg N, Mars N, Thorn LM, Harjutsalo V, Rossing P, Ahluwalia TS, Lindsberg PJ, Sandholm N, Groop PH. The Low-Expression Variant of FABP4 Is Associated With Cardiovascular Disease in Type 1 Diabetes. Diabetes 2021; 70:2391-2401. [PMID: 34244239 DOI: 10.2337/db21-0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022]
Abstract
Fatty acid binding protein 4 (FABP4) is implicated in the pathogenesis of cardiometabolic disorders. Pharmacological inhibition or genetic deletion of FABP4 improves cardiometabolic health and protects against atherosclerosis in preclinical models. As cardiovascular disease (CVD) is common in type 1 diabetes, we examined the role of FABP4 in the development of complications in type 1 diabetes, focusing on a functional, low-expression variant (rs77878271) in the promoter of the FABP4 gene. For this, we assessed the risk of CVD, stroke, coronary artery disease (CAD), end-stage kidney disease, and mortality using Cox proportional hazards models for the FABP4 rs77878271 in 5,077 Finnish individuals with type 1 diabetes. The low-expression G allele of rs77878271 increased the risk of CVD, independent of confounders. Findings were tested for replication in 852 Danish and 3,678 Finnish individuals with type 1 diabetes. In the meta-analysis, each G allele increased the risk of stroke by 26% (P = 0.04), CAD by 26% (P = 0.006), and CVD by 17% (P = 0.003). In Mendelian randomization, a 1-SD unit decrease in FABP4 increased risk of CAD 2.4-fold. Hence, in contrast with the general population, among patients with type 1 diabetes the low-expression G allele of rs77878271 increased CVD risk, suggesting that genetically low FABP4 levels may be detrimental in the context of type 1 diabetes.
Collapse
Affiliation(s)
- Emma H Dahlström
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani Saksi
- Neurology, Neurocenter, Helsinki University Hospital, and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Nina Mars
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Lena M Thorn
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Perttu J Lindsberg
- Neurology, Neurocenter, Helsinki University Hospital, and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Niina Sandholm
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
20
|
Adipose-Derived Lipid-Binding Proteins: The Good, the Bad and the Metabolic Diseases. Int J Mol Sci 2021; 22:ijms221910460. [PMID: 34638803 PMCID: PMC8508731 DOI: 10.3390/ijms221910460] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue releases a large range of bioactive factors called adipokines, many of which are involved in inflammation, glucose homeostasis and lipid metabolism. Under pathological conditions such as obesity, most of the adipokines are upregulated and considered as deleterious, due to their pro-inflammatory, pro-atherosclerotic or pro-diabetic properties, while only a few are downregulated and would be designated as beneficial adipokines, thanks to their counteracting properties against the onset of comorbidities. This review focuses on six adipose-derived lipid-binding proteins that have emerged as key factors in the development of obesity and diabetes: Retinol binding protein 4 (RBP4), Fatty acid binding protein 4 (FABP4), Apolipoprotein D (APOD), Lipocalin-2 (LCN2), Lipocalin-14 (LCN14) and Apolipoprotein M (APOM). These proteins share structural homology and capacity to bind small hydrophobic molecules but display opposite effects on glucose and lipid metabolism. RBP4 and FABP4 are positively associated with metabolic syndrome, while APOD and LCN2 are ubiquitously expressed proteins with deleterious or beneficial effects, depending on their anatomical site of expression. LCN14 and APOM have been recently identified as adipokines associated with healthy metabolism. Recent findings on these lipid-binding proteins exhibiting detrimental or protective roles in human and murine metabolism and their involvement in metabolic diseases are also discussed.
Collapse
|