1
|
Zhang J, Xu Q, Sun G. Lipocalin-2 promotes NSCLC progression by activating the JAK2/STAT3 signaling pathway. J Transl Med 2025; 23:419. [PMID: 40211270 PMCID: PMC11987316 DOI: 10.1186/s12967-025-06418-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Lipocalin-2 (LCN2), a pleiotropic protein implicated in tumorigenesis and cancer progression, has been associated with multiple malignancies. However, its precise role in NSCLC and the underlying molecular mechanisms remain incompletely understood. This study aimed to elucidate the function of LCN2 in NSCLC, with a particular focus on its involvement in the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway. METHODS LCN2 expression in NSCLC tissues was comprehensively analyzed using bioinformatics tools, including the Universal Analysis of Cancer (UALCAN), The Cancer Genome Atlas (TCGA), UCSC-XENA, and Gene Expression Omnibus (GEO) databases. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were employed to assess LCN2 expression levels in NSCLC cell lines. The functional impact of LCN2 on NSCLC cells, including proliferation, apoptosis, and metastasis, were assessed through a series of in vitro assays, such as Cell Counting Kit-8 (CCK-8), EdU, wound healing, and transwell migration and invasion assays. An in vivo xenograft model was established to investigate the effects of LCN2 on tumor growth and metastasis. Additionally, the involvement of the JAK2/STAT3 signaling pathway was examined using western blotting and pharmacological inhibition with AG490. RESULTS LCN2 was significantly upregulated in NSCLC tissues and cell lines, and its elevated expression correlated with poor prognosis. Functional analyses demonstrated that LCN2 knockdown suppressed NSCLC cell proliferation, migration, and invasion while promoting apoptosis. Mechanistically, LCN2 was found to activate the JAK2/STAT3 pathway by interacting with SOCS3, and pharmacological blockade of this pathway effectively abrogated the oncogenic effects of LCN2 overexpression. CONCLUSIONS This study identifies LCN2 as a potential oncogene in NSCLC, driving tumor progression through activation of the JAK2/STAT3 signaling pathway. These findings suggest that targeting LCN2 or its downstream signaling components may represent a promising therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Province, 230022, Hefei, China
| | - Qin Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Province, 230022, Hefei, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Province, 230022, Hefei, China.
| |
Collapse
|
2
|
Bauer C, Patten RK, Sun Q, Li H, Konja D, Woessner M, Lin X, Garnham A, Hare DL, Ebeling PR, Sim M, Lewis JR, Wang Y, Parker L, Levinger I. The effect of prednisolone ingestion and acute exercise on lipocalin-2 and its variants in young men: a pilot randomised crossover study. Sci Rep 2025; 15:4453. [PMID: 39910157 PMCID: PMC11799433 DOI: 10.1038/s41598-025-88115-z] [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: 04/24/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Lipocalin-2 (LCN2) has three main variants; polyaminated (hLCN2) and non-polyaminated (C87A and R81E). The polyaminated form is proposed to positively influence energy control, whereas the non-polyaminated forms negatively impact energy control in mice. Glucocorticoids negatively affect glucose regulation and exercise has a positive effect. We hypothesise that glucocorticoids will suppress, while exercise will increase hLCN2, and decrease C87A and R81E, which will be associated with improved insulin sensitivity. In a randomised crossover design, nine young healthy men (aged 27.8 ± 4.9 years; BMI 24.4 ± 2.4 kg/m2) completed 30 min of high-intensity aerobic exercise (90-95% heart rate reserve) after glucocorticoid or placebo ingestion. Blood was collected and analyzed for LCN2 and its variants levels at baseline, immediately, 60 min and 180 min post-exercise. Insulin sensitivity was assessed using hyperinsulinemic-euglycemic clamp. A main effect, increase in LCN2 was detected for prednisolone ingestion (overall treatment effect p = 0.001), but not LCN2 variants (all p > 0.05). Main effects for time were observed for exercise for LCN2 and all variants (overall time effect all p < 0.02). Regardless of treatment, LCN2, C87A, R81E, and hLCN2 increased immediately after exercise compared with baseline (all p < 0.04). C87A, but not LCN2 or its other variants, remained elevated at 180 min post-ex (p = 0.007). LCN2, but not its variants, was elevated in response to prednisolone ingestion. LCN2 and its variants are transiently increased by acute exercise, but this increase was not related to insulin sensitivity. The clinical implication of elevated LCN2 and its variants post-exercise on satiety and energy regulation, as well as the mechanisms involved warrant further investigation.Clinical trial registration: www.anzctr.org.au , ACTRN12615000755538.
Collapse
Affiliation(s)
- Carlie Bauer
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, 3011, Australia
| | - Rhiannon K Patten
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, 3011, Australia
| | - Qiyang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Haoyun Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mary Woessner
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, 3011, Australia
| | - Xuzhu Lin
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Andrew Garnham
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, 3011, Australia
| | - David L Hare
- Department of Cardiology, Austin Health, University of Melbourne, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
- Department of Endocrinology, Monash Health, Clayton, VIC, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia
- Medical School, The University of Western Australia, Perth, Australia
| | - Joshua R Lewis
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Joondalup, Australia
- Medical School, The University of Western Australia, Perth, Australia
- Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, 3011, Australia.
- Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, Western Health, St. Albans, Australia.
| |
Collapse
|
3
|
Zhang Y, Song X, Qi T, Gao S, Sun C, Yang J, Zhou X. Association between lipocalin-2 levels and diabetic retinopathy in patients with overweight/obese type 2 diabetes mellitus. Ir J Med Sci 2023; 192:2785-2792. [PMID: 37069380 DOI: 10.1007/s11845-023-03365-y] [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: 10/15/2022] [Accepted: 04/03/2023] [Indexed: 04/19/2023]
Abstract
AIMS The study aimed to investigate the association between lipocalin-2 (LCN-2) levels and diabetic retinopathy (DR) in patients with overweight/obese type 2 diabetes mellitus(T2DM), and to explore the mechanism of LCN-2 in overweight/obese DR. METHODS The study involved 237 T2DM inpatients divided into the normal group and overweight/obese group, and the two groups were further divided into two subgroups according to the presence or absence of DR. The demographic data and biochemical parameters were measured. RESULTS LCN-2 levels in overweight/obese groups were higher than those in normal groups (P < 0.001 for all), and patients with DR had higher levels of LCN-2 than those without DR(P < 0.05 for all) in normal groups and overweight/obese groups. Binary logistic regression analysis showed that no significant significance was observed for LCN-2 levels compared to those below the median in the normal group, but individuals with LCN-2 levels above the median had 4.198 times higher risk of developing DR than those below the median (OR = 4.198, 95% CI = 1.676-10.516) after adjustment for potential confounding factors in the overweight/obese group. In the total, normal and overweight/obese groups, the prediction capacity of LCN-2 for DR was 1.56, 1.58 and 1.65 times, respectively. Conclusionsː In conclusion, our study found that LCN-2 levels were higher in overweight/obese patients with DR, and LCN-2 was an independent predictor of DR in T2DM patients with overweight/obese. In addition, LCN-2 may be a valuable predictor of DR-like factors such as the duration of diabetes and hypertension.
Collapse
Affiliation(s)
- Yajuan Zhang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Xiaojun Song
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Tianying Qi
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Shan Gao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China
| | - Chao Sun
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jiaxuan Yang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xinli Zhou
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China.
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China.
| |
Collapse
|
4
|
Sciarretta F, Ceci V, Tiberi M, Zaccaria F, Li H, Zhou ZY, Sun Q, Konja D, Matteocci A, Bhusal A, Verri M, Fresegna D, Balletta S, Ninni A, Di Biagio C, Rosina M, Suk K, Centonze D, Wang Y, Chiurchiù V, Aquilano K, Lettieri-Barbato D. Lipocalin-2 promotes adipose-macrophage interactions to shape peripheral and central inflammatory responses in experimental autoimmune encephalomyelitis. Mol Metab 2023; 76:101783. [PMID: 37517520 PMCID: PMC10448472 DOI: 10.1016/j.molmet.2023.101783] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
OBJECTIVE Accumulating evidence suggests that dysfunctional adipose tissue (AT) plays a major role in the risk of developing multiple sclerosis (MS), the most common immune-mediated and demyelinating disease of the central nervous system. However, the contribution of adipose tissue to the etiology and progression of MS is still obscure. This study aimed at deciphering the responses of AT in experimental autoimmune encephalomyelitis (EAE), the best characterized animal model of MS. RESULTS AND METHODS We observed a significant AT loss in EAE mice at the onset of disease, with a significant infiltration of M1-like macrophages and fibrosis in the AT, resembling a cachectic phenotype. Through an integrative and multilayered approach, we identified lipocalin2 (LCN2) as the key molecule released by dysfunctional adipocytes through redox-dependent mechanism. Adipose-derived LCN2 shapes the pro-inflammatory macrophage phenotype, and the genetic deficiency of LCN2 specifically in AT reduced weight loss as well as inflammatory macrophage infiltration in spinal cord in EAE mice. Mature adipocytes downregulating LCN2 reduced lipolytic response to inflammatory stimuli (e.g. TNFα) through an ATGL-mediated mechanism. CONCLUSIONS Overall data highlighted a role LCN2 in exacerbating inflammatory phenotype in EAE model, suggesting a pathogenic role of dysfunctional AT in MS.
Collapse
Affiliation(s)
| | - Veronica Ceci
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Fabio Zaccaria
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Haoyun Li
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhong-Yan Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiyang Sun
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Martina Verri
- Pathology Unit, University Hospital Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00163 Rome, Italy
| | - Sara Balletta
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Andrea Ninni
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Di Biagio
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marco Rosina
- Neurology Unit, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daniele Lettieri-Barbato
- IRCCS, Fondazione Santa Lucia, 00179 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
5
|
Patel S, Sparman NZR, Arneson D, Alvarsson A, Santos LC, Duesman SJ, Centonze A, Hathaway E, Ahn IS, Diamante G, Cely I, Cho CH, Talari NK, Rajbhandari AK, Goedeke L, Wang P, Butte AJ, Blanpain C, Chella Krishnan K, Lusis AJ, Stanley SA, Yang X, Rajbhandari P. Mammary duct luminal epithelium controls adipocyte thermogenic programme. Nature 2023; 620:192-199. [PMID: 37495690 PMCID: PMC10529063 DOI: 10.1038/s41586-023-06361-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
Sympathetic activation during cold exposure increases adipocyte thermogenesis via the expression of mitochondrial protein uncoupling protein 1 (UCP1)1. The propensity of adipocytes to express UCP1 is under a critical influence of the adipose microenvironment and varies between sexes and among various fat depots2-7. Here we report that mammary gland ductal epithelial cells in the adipose niche regulate cold-induced adipocyte UCP1 expression in female mouse subcutaneous white adipose tissue (scWAT). Single-cell RNA sequencing shows that glandular luminal epithelium subtypes express transcripts that encode secretory factors controlling adipocyte UCP1 expression under cold conditions. We term these luminal epithelium secretory factors 'mammokines'. Using 3D visualization of whole-tissue immunofluorescence, we reveal sympathetic nerve-ductal contact points. We show that mammary ducts activated by sympathetic nerves limit adipocyte UCP1 expression via the mammokine lipocalin 2. In vivo and ex vivo ablation of mammary duct epithelium enhance the cold-induced adipocyte thermogenic gene programme in scWAT. Since the mammary duct network extends throughout most of the scWAT in female mice, females show markedly less scWAT UCP1 expression, fat oxidation, energy expenditure and subcutaneous fat mass loss compared with male mice, implicating sex-specific roles of mammokines in adipose thermogenesis. These results reveal a role of sympathetic nerve-activated glandular epithelium in adipocyte UCP1 expression and suggest that mammary duct luminal epithelium has an important role in controlling glandular adiposity.
Collapse
Affiliation(s)
- Sanil Patel
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Njeri Z R Sparman
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Arneson
- Department of Integrative Biology and Physiology and Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luís C Santos
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samuel J Duesman
- Department of Psychiatry and Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessia Centonze
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ephraim Hathaway
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology and Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology and Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Ingrid Cely
- Department of Integrative Biology and Physiology and Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Chung Hwan Cho
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noble Kumar Talari
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Abha K Rajbhandari
- Department of Psychiatry and Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leigh Goedeke
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peng Wang
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
- Center for Data-Driven Insights and Innovation, University of California Health, Oakland, CA, USA
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Medicine, Division of Cardiology, and Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, and Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology and Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Wang D, Li X, Jiao D, Cai Y, Qian L, Shen Y, Lu Y, Zhou Y, Fu B, Sun R, Tian Z, Zheng X, Wei H. LCN2 secreted by tissue-infiltrating neutrophils induces the ferroptosis and wasting of adipose and muscle tissues in lung cancer cachexia. J Hematol Oncol 2023; 16:30. [PMID: 36973755 PMCID: PMC10044814 DOI: 10.1186/s13045-023-01429-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/19/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Cancer cachexia is a deadly wasting syndrome that accompanies various diseases (including ~ 50% of cancers). Clinical studies have established that cachexia is not a nutritional deficiency and is linked to expression of certain proteins (e.g., interleukin-6 and C-reactive protein), but much remains unknown about this often fatal syndrome. METHODS First, cachexia was created in experimental mouse models of lung cancer. Samples of human lung cancer were used to identify the association between the serum lipocalin 2 (LCN2) level and cachexia progression. Then, mouse models with LCN2 blockade or LCN2 overexpression were used to ascertain the role of LCN2 upon ferroptosis and cachexia. Furthermore, antibody depletion of tissue-infiltrating neutrophils (TI-Neu), as well as myeloid-specific-knockout of Lcn2, were undertaken to reveal if LCN2 secreted by TI-Neu caused cachexia. Finally, chemical inhibition of ferroptosis was conducted to illustrate the effect of ferroptosis upon tissue wasting. RESULTS Protein expression of LCN2 was higher in the wasting adipose tissue and muscle tissues of experimental mouse models of lung cancer cachexia. Moreover, evaluation of lung cancer patients revealed an association between the serum LCN2 level and cachexia progression. Inhibition of LCN2 expression reduced cachexia symptoms significantly and inhibited tissue wasting in vivo. Strikingly, we discovered a significant increase in the number of TI-Neu in wasting tissues, and that these innate immune cells secreted high levels of LCN2. Antibody depletion of TI-Neu, as well as myeloid-specific-knockout of Lcn2, prevented ferroptosis and tissue wasting in experimental models of lung cancer cachexia. Chemical inhibition of ferroptosis alleviated tissue wasting significantly and also prolonged the survival of cachectic mice. CONCLUSIONS Our study provides new insights into how LCN2-induced ferroptosis functionally impacts tissue wasting. We identified LCN2 as a potential target in the treatment of cancer cachexia.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Xiaohui Li
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Defeng Jiao
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Ying Cai
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Liting Qian
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yiqing Shen
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yichen Lu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yonggang Zhou
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Binqing Fu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Rui Sun
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Zhigang Tian
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Xiaohu Zheng
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Haiming Wei
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Institue of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
7
|
Investigation of the multi-targeted protection potential of tannic acid against doxorubicin-induced kidney damage in rats. Chem Biol Interact 2022; 365:110111. [PMID: 35987278 DOI: 10.1016/j.cbi.2022.110111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022]
Abstract
Doxorubicin (DOX) is an antitumor drug that is powerful but can cause worse outcomes, including nephrotoxicity, and therefore has limited clinical use. Therefore, it is necessary to identify safer agents that can minimize the damage caused by the drug without shifting the treatment performance, in addition to clarifying the underlying mechanisms of DOX-induced aberrant in vivo renal activation. In this study, we tested the prophylactic capacity and mechanisms of action of tannic acid (TA) against DOX-mediated kidney damage in rats and evaluated the nephrotoxic activity of DOX when used with TA. Rats were treated during the two weeks with cumulative (18 mg/kg with six different injections) DOX, daily TA (50 mg/kg), and the DOX + TA combination. Changes in major metabolites and components involved in antioxidant metabolism were evaluated in the kidney tissues of all animals. Further, the gene expression levels of regulatory factors that have critical importance in cell metabolism, inflammation, and apoptosis were investigated. Both biochemical and molecular examinations showed that TA improved DOX-induced dysregulations at both protein and gene levels in the kidneys. Increased lipid peroxidation and decreased glutathione levels were reversed. Consistent with oxidative stress marker metabolites, suppressed antioxidant enzyme activities and transcript levels of antioxidant system members were restored. Of note, combination treatment with TA could overcome doxorubicin-induced gene expressions markedly altered by DOX, suggesting that nephroprotection conferred by TA involved the remodeling of stress resistance, cell metabolism, inflammation, and apoptosis. Collectively, the present in vivo study suggests that TA could be used as a multitarget and effective agent for the mitigation of doxorubicin-induced nephrotoxicity without changing the therapeutic efficacy of the drug.
Collapse
|
8
|
Owen A, Patel JM, Parekh D, Bangash MN. Mechanisms of Post-critical Illness Cardiovascular Disease. Front Cardiovasc Med 2022; 9:854421. [PMID: 35911546 PMCID: PMC9334745 DOI: 10.3389/fcvm.2022.854421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged critical care stays commonly follow trauma, severe burn injury, sepsis, ARDS, and complications of major surgery. Although patients leave critical care following homeostatic recovery, significant additional diseases affect these patients during and beyond the convalescent phase. New cardiovascular and renal disease is commonly seen and roughly one third of all deaths in the year following discharge from critical care may come from this cluster of diseases. During prolonged critical care stays, the immunometabolic, inflammatory and neurohumoral response to severe illness in conjunction with resuscitative treatments primes the immune system and parenchymal tissues to develop a long-lived pro-inflammatory and immunosenescent state. This state is perpetuated by persistent Toll-like receptor signaling, free radical mediated isolevuglandin protein adduct formation and presentation by antigen presenting cells, abnormal circulating HDL and LDL isoforms, redox and metabolite mediated epigenetic reprogramming of the innate immune arm (trained immunity), and the development of immunosenescence through T-cell exhaustion/anergy through epigenetic modification of the T-cell genome. Under this state, tissue remodeling in the vascular, cardiac, and renal parenchymal beds occurs through the activation of pro-fibrotic cellular signaling pathways, causing vascular dysfunction and atherosclerosis, adverse cardiac remodeling and dysfunction, and proteinuria and accelerated chronic kidney disease.
Collapse
Affiliation(s)
- Andrew Owen
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jaimin M. Patel
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Mansoor N. Bangash
- Department of Critical Care, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, United Kingdom
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Mansoor N. Bangash
| |
Collapse
|
9
|
Nakamura T, Bonnard B, Palacios-Ramirez R, Fernández-Celis A, Jaisser F, López-Andrés N. Biglycan Is a Novel Mineralocorticoid Receptor Target Involved in Aldosterone/Salt-Induced Glomerular Injury. Int J Mol Sci 2022; 23:ijms23126680. [PMID: 35743123 PMCID: PMC9224513 DOI: 10.3390/ijms23126680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 11/16/2022] Open
Abstract
The beneficial effects of mineralocorticoid receptor (MR) antagonists (MRAs) for various kidney diseases are established. However, the underlying mechanisms of kidney injury induced by MR activation remain to be elucidated. We recently reported aldosterone-induced enhancement of proteoglycan expression in mitral valve interstitial cells and its association with fibromyxomatous valvular disorder. As the expression of certain proteoglycans is elevated in several kidney diseases, we hypothesized that proteoglycans mediate kidney injury in the context of aldosterone/MR pathway activation. We evaluated the proteoglycan expression and tissue injury in the kidney and isolated glomeruli of uninephrectomy/aldosterone/salt (NAS) mice. The MRA eplerenone was administered to assess the role of the MR pathway. We investigated the direct effects of biglycan, one of the proteoglycans, on macrophages using isolated macrophages. The kidney samples from NAS-treated mice showed enhanced fibrosis and increased expression of biglycan accompanying glomerular macrophage infiltration and enhanced expression of TNF-α, iNOS, Nox2, CCL3 (C-C motif chemokine ligand 3), and phosphorylated NF-κB. Eplerenone blunted these changes. Purified biglycan stimulated macrophages to express TNF-α, iNOS, Nox2, and CCL3. This was prevented by a toll-like receptor 4 (TLR4) or NF-κB inhibitor, indicating that biglycan stimulation is dependent on the TLR4/NF-κB pathway. We identified the proteoglycan biglycan as a novel target of MR involved in MR-induced glomerular injury and macrophage infiltration via a biglycan/TLR4/NF-κB/CCL3 cascade.
Collapse
Affiliation(s)
- Toshifumi Nakamura
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Roberto Palacios-Ramirez
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
- INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), 54500 Nancy, France
- Correspondence: (F.J.); (N.L.-A.); Tel.: +33-144276485 (F.J.); +34-848428539 (N.L.-A.)
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (F.J.); (N.L.-A.); Tel.: +33-144276485 (F.J.); +34-848428539 (N.L.-A.)
| |
Collapse
|
10
|
Ren K, Xia Y. Lipocalin 2 Participates in the Epidermal Differentiation and Inflammatory Processes of Psoriasis. J Inflamm Res 2022; 15:2157-2166. [PMID: 35386225 PMCID: PMC8979418 DOI: 10.2147/jir.s358492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
As a multifunctional cytokine, lipocalin 2 is weakly expressed in skin and serum under normal conditions. However, it is over-expressed by neutrophils and keratinocytes in the skin lesions and sera in several skin diseases. Recent studies demonstrated that lipocalin 2 participates in the pathogenesis of psoriasis by exerting versatile effects on skin resident cells and infiltrating immune cells. Lipocalin 2 inhibits the synthesis of keratin, involucrin, and loricrin in keratinocytes, leading to epidermal parakeratosis via the Tcf7l1-lipocalin 2 signaling axis. It also recruits inflammatory cells such as T cells and neutrophils into skin lesions via the IL-23/IL17, p38-MAPK, and ERK-1/2 signaling pathways. Additionally, lipocalin 2 and other cytokines such as IL-17 have the synergetic effects on skin cells. The neutralization of lipocalin 2 or relevant cytokines can alleviate psoriasis, verifying that lipocalin 2 is an effective interfering target for psoriasis. In this review, we summarize the roles of lipocalin 2 in the processes of psoriatic inflammation and the promising therapeutic strategies based on lipocalin 2-related molecules.
Collapse
Affiliation(s)
- Kaixuan Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
11
|
Vanarsa K, Sasidharan P, Duran V, Gokaraju S, Nidhi M, Louis Sam Titus ASC, Soomro S, Stock AD, Der E, Putterman C, Greenberg B, Mok CC, Hanly JG, Mohan C. Aptamer-based screen of Neuropsychiatric Lupus cerebrospinal fluid reveals potential biomarkers that overlap with the choroid plexus transcriptome. Arthritis Rheumatol 2022; 74:1223-1234. [PMID: 35099126 DOI: 10.1002/art.42080] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES As no gold-standard diagnostic test exists for neuropsychiatric systemic lupus erythematosus (NPSLE), we executed a broad screen of NPSLE cerebrospinal fluid (CSF) using an aptamer-based platform. METHODS CSF were obtained from NPSLE patients and subjected to proteomic assay using the aptamer-based screen. Potential biomarkers were identified and validated in independent NPSLE cohorts in comparison with other neurological diseases. RESULTS 40 proteins out of 1129 screened were elevated in NPSLE CSF. By ELISA validation, CSF Angiostatin, α2-Macroglobulin, DAN, Fibronectin, HCC-1, IgM, Lipocalin 2, M-CSF and SERPING1 were significantly elevated in a predominantly Caucasian NPSLE cohort (n=24), compared to patients with other neurological diseases (n=54), with CSF IgM (AUC=0.95) and M-CSF (AUC=0.91) being the most discriminatory. In a second, Hong Kong NPSLE cohort, CSF IgM (AUC=0.78) and Lipocalin-2 (AUC=0.85) were the most discriminatory. Several CSF proteins exhibited high diagnostic specificity for NPSLE in both cohorts. Elevated CSF C3 was associated with acute confusional state. Eleven molecules elevated in NPSLE CSF exhibited concordant elevation in the choroid plexus, suggesting shared origins. CONCLUSIONS CSF Lipocalin-2, M-CSF, IgM and complement C3 emerge as promising CSF biomarkers of NPSLE with diagnostic potential.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Valeria Duran
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Sirisha Gokaraju
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Malavika Nidhi
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Evan Der
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | - John G Hanly
- Division of Rheumatology, Queen Elizabeth II Health Sciences Center and Dalhousie University Halifax, Nova Scotia, Canada
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX
| |
Collapse
|
12
|
Sanz-Lamora H, Marrero PF, Haro D, Relat J. A Mixture of Pure, Isolated Polyphenols Worsens the Insulin Resistance and Induces Kidney and Liver Fibrosis Markers in Diet-Induced Obese Mice. Antioxidants (Basel) 2022; 11:120. [PMID: 35052623 PMCID: PMC8772794 DOI: 10.3390/antiox11010120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a worldwide epidemic with severe metabolic consequences. Polyphenols are secondary metabolites in plants and the most abundant dietary antioxidants, which possess a wide range of health effects. The most relevant food sources are fruit and vegetables, red wine, black and green tea, coffee, virgin olive oil, and chocolate, as well as nuts, seeds, herbs, and spices. The aim of this work was to evaluate the ability of a pure, isolated polyphenol supplementation to counteract the pernicious metabolic effects of a high-fat diet (HFD). Our results indicated that the administration of pure, isolated polyphenols under HFD conditions for 26 weeks worsened the glucose metabolism in diet-induced obese mice. The data showed that the main target organ for these undesirable effects were the kidneys, where we observed fibrotic, oxidative, and kidney-disease markers. This work led us to conclude that the administration of pure polyphenols as a food supplement would not be advisable. Instead, the ingestion of complete "whole" foods would be the best way to get the health effects of bioactive compounds such as polyphenols.
Collapse
Affiliation(s)
- Hèctor Sanz-Lamora
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute for Nutrition and Food Safety Research, University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
| | - Pedro F. Marrero
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute of Biomedicine, University of Barcelona (IBUB), E-08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Diego Haro
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute of Biomedicine, University of Barcelona (IBUB), E-08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute for Nutrition and Food Safety Research, University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
13
|
Carvajal CA, Tapia-Castillo A, Pérez JA, Fardella CE. Serum Alpha-1-Acid Glycoprotein-1 and Urinary Extracellular Vesicle miR-21-5p as Potential Biomarkers of Primary Aldosteronism. Front Immunol 2021; 12:768734. [PMID: 34804057 PMCID: PMC8603108 DOI: 10.3389/fimmu.2021.768734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Primary aldosteronism (PA) is the most common cause of secondary hypertension and reaches a prevalence of 6-10%. PA is an endocrine disorder, currently identified as a broad-spectrum phenotype, spanning from normotension to hypertension. In this regard, several studies have made advances in the identification of mediators and novel biomarkers of PA as specific proteins, miRNAs, and lately, extracellular vesicles (EVs) and their cargo. Aim To evaluate lipocalins LCN2 and AGP1, and specific urinary EV miR-21-5p and Let-7i-5p as novel biomarkers for PA. Subjects and Methods A cross-sectional study was performed in 41 adult subjects classified as normotensive controls (CTL), essential hypertensives (EH), and primary aldosteronism (PA) subjects, who were similar in gender, age, and BMI. Systolic (SBP) and diastolic (DBP) blood pressure, aldosterone, plasma renin activity (PRA), and aldosterone to renin ratio (ARR) were determined. Inflammatory parameters were defined as hs-C-reactive protein (hs-CRP), PAI-1, MMP9, IL6, LCN2, LCN2-MMP9, and AGP1. We isolated urinary EVs (uEVs) and measured two miRNA cargo miR-21-5p and Let-7i-5p by Taqman-qPCR. Statistical analyses as group comparisons were performed by Kruskall-Wallis, and discriminatory analyses by ROC curves were performed with SPSS v21 and Graphpad-Prism v9. Results PA and EH subjects have significantly higher SBP and DBP (p <0.05) than the control group. PA subjects have similar hs-CRP, PAI-1, IL-6, MMP9, LCN2, and LCN2-MMP9 but have higher levels of AGP1 (p <0.05) than the CTL&EH group. The concentration and size of uEVs and miRNA Let-7i-5p did not show any difference between groups. In PA, we found significantly lower levels of miR-21-5p than controls (p <0.05). AGP1 was associated with aldosterone, PRA, and ARR. ROC curves detected AUC for AGP1 of 0.90 (IC 95 [0.79 - 1.00], p <0.001), and combination of AGP1 and EV-miR-21-5p showed an AUC of 0.94 (IC 95 [0.85 - 1.00], p<0.001) to discriminate the PA condition from EH and controls. Conclusion Serum AGP1 protein was found to be increased, and miR-21-5p in uEVs was decreased in subjects classified as PA. Association of AGP1 with aldosterone, renin activity, and ARR, besides the high discriminatory capacity of AGP1 and uEV-miR-21-5p to identify the PA condition, place both as potential biomarkers of PA.
Collapse
Affiliation(s)
- Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Pérez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
14
|
Bonnard B, Ibarrola J, Lima-Posada I, Fernández-Celis A, Durand M, Genty M, Lopez-Andreés N, Jaisser F. Neutrophil Gelatinase-Associated Lipocalin From Macrophages Plays a Critical Role in Renal Fibrosis Via the CCL5 (Chemokine Ligand 5)-Th2 Cells-IL4 (Interleukin 4) Pathway. Hypertension 2021; 79:352-364. [PMID: 34794340 DOI: 10.1161/hypertensionaha.121.17712] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NGAL (neutrophil gelatinase-associated lipocalin; or lipocalin 2, Lcn2) is a novel mineralocorticoid target in the cardiovascular system. We showed that Lcn2 gene invalidation protects against proteinuria and renal injury upon mineralocorticoid excess and we hypothesized that NGAL produced from macrophages promotes the expression of chemoattractant molecules involved these renal lesions. The role of NGAL was analyzed using myeloid-specific (MΦ KO NGAL) Lcn2 knockout mice challenged with uni-nephrectomy, aldosterone, and salt (NAS) for 6 weeks. The role of the CCL5 (chemokine ligand 5) and IL4 (interleukin 4) in kidney fibrosis was studied by administration of the CCL5 receptor antagonist maraviroc or by injections of an anti-IL4 neutralizing antibody. In CTL mice, NAS increased the renal expression of extracellular matrix proteins, such as collagen I, αSMA, and fibronectin associated with interstitial fibrosis which were blunted in MΦ KO NGAL mice. The expression of CCL5 was blunted in sorted macrophages from MΦ KO NGAL mice challenged by NAS and in macrophages obtained from KO NGAL mice and challenged ex vivo with aldosterone and salt. The pharmacological blockade of the CCL5 receptor reduced renal fibrosis and the CD4+ Th cell infiltration induced by NAS. Neutralization of IL4 in NAS mice blunted kidney fibrosis and the overexpression of profibrotic proteins, such as collagen I, αSMA, and fibronectin. In conclusion, NGAL produced by macrophages plays a critical role in renal fibrosis and modulates the CCL5/IL4 pathway in mice exposed to mineralocorticoid excess.
Collapse
Affiliation(s)
- Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Jaime Ibarrola
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Ixchel Lima-Posada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Manon Durand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Marie Genty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Natalia Lopez-Andreés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.).,INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France (F.J.)
| |
Collapse
|
15
|
Li D, Li H, Bauer C, Hu Y, Lewis JR, Xu A, Levinger I, Wang Y. Lipocalin-2 Variants and Their Relationship With Cardio-Renal Risk Factors. Front Endocrinol (Lausanne) 2021; 12:781763. [PMID: 34938273 PMCID: PMC8685543 DOI: 10.3389/fendo.2021.781763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/09/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES To investigate the serum, plasma and urine levels of lipocalin-2 (LCN2) variants in healthy humans and their associations with risk factors for cardiometabolic (CMD) and chronic kidney (CKD) diseases. METHODS Fifty-nine males and 41 females participated in the study. Blood and urine were collected following an overnight fasting. LCN2 variants were analyzed using validated in-house ELISA kits. Heart rate, blood pressure, lipids profile, glucose, adiponectin, high-sensitivity C-reactive protein (hsCRP), creatinine, cystatin C, and biomarkers for kidney function were assessed. RESULTS The levels of hLcn2, C87A and R81E in serum and urine, but not plasma, were significantly higher in men than women. Increased levels of LCN2 variants, as well as their relative ratios, in serum and plasma were positively associated with body mass index, blood pressure, triglyceride and hsCRP (P<0.05). No significant correlations were found between these measures and hLcn2, C87A or R81E in urine. However, LCN2 variants in urine, but not plasma or serum, were correlated with biomarkers of kidney function (P<0.05). CONCLUSIONS Both the serum and plasma levels of LCN2 variants, as well as their ratios are associated with increased cardiometabolic risk, whereas those in urine are correlated with renal dysfunction. LCN2 variants represent promising biomarkers for CMD and CKD.
Collapse
Affiliation(s)
- Dahui Li
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Haoyun Li
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Carlie Bauer
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Yue Hu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Joshua R. Lewis
- Medical School, University of Western Australia, Perth, WA, Australia
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Centre for Kidney Research, Children’s Hospital at Westmead, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Aimin Xu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, University of Melbourne, Western Health, St. Albans, VIC, Australia
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Yu Wang,
| |
Collapse
|
16
|
Antimicrobial peptides: bridging innate and adaptive immunity in the pathogenesis of psoriasis. Chin Med J (Engl) 2020; 133:2966-2975. [PMID: 33237697 PMCID: PMC7752697 DOI: 10.1097/cm9.0000000000001240] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are small molecules produced by a myriad of cells and play important roles not only in protecting against infections and sustaining skin barrier homeostasis but also in contributing to immune dysregulation under pathological conditions. Recently, increasing evidence has indicated that AMPs, including cathelicidin (LL-37), human β-defensins, S100 proteins, lipocalin 2, and RNase 7, are highly expressed in psoriatic skin lesions. These peptides broadly regulate immunity by interacting with various immune cells and linking innate and adaptive immune responses during the progression of psoriasis. In this review, we summarize the recent findings regarding AMPs in the pathogenesis of psoriasis with a main focus on their immunomodulatory abilities.
Collapse
|
17
|
Zhang P, Konja D, Wang Y. Adipose tissue secretory profile and cardiometabolic risk in obesity. ENDOCRINE AND METABOLIC SCIENCE 2020. [DOI: 10.1016/j.endmts.2020.100061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
18
|
Ong KL, Wu L, Januszewski AS, O'Connell RL, Xu A, Rye KA, Ma RCW, Li H, Jenkins AJ, Jia W, Keech AC. Relationships of adipocyte-fatty acid binding protein and lipocalin 2 with risk factors and chronic complications in type 2 diabetes and effects of fenofibrate: A fenofibrate Intervention and event lowering in diabetes sub-study. Diabetes Res Clin Pract 2020; 169:108450. [PMID: 32949655 DOI: 10.1016/j.diabres.2020.108450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/24/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
AIMS To investigate determinants of circulating levels of adipocyte-fatty acid binding protein (A-FABP) and lipocalin-2 (LCN2), their relationships with cardiovascular disease (CVD) and microvascular events, and effects of fenofibrate in type 2 diabetes (T2D). METHODS A-FABP and LCN2 were quantified in baseline plasma from 2000 T2D adults in a Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) trial sub-study and correlates thereof determined. In a subset (n = 200) adipokines were also measured on-trial. RESULTS Female sex, older age, higher body mass index (BMI), HbA1c, insulin resistance index, triglycerides, plasma creatinine and homocysteine, shorter diabetes duration, and use of oral hypoglycaemic agents alone were independent determinants of higher A-FABP. Higher BMI, fibrinogen and homocysteine, Caucasian race, and lower fasting glucose, HDL-cholesterol, apolipoprotein A-II and estimated glomerular filtration rate were independent predictors of higher LCN2 levels. Baseline A-FABP and LCN2 levels were associated with multiple new CVD and microvascular events over 5-years, though significance was lost after risk factor adjustment. Fenofibrate increased A-FABP but did not change LCN2 levels. CONCLUSIONS Baseline plasma A-FABP and LCN2 levels were associated with concurrent CVD risk factors, and on-trial chronic complications, likely mediated via traditional risk factors. Fenofibrate increased A-FABP modestly but did not affect LCN2 levels. CLINICAL TRIAL REGISTRATION ISRCTN 64783481.
Collapse
Affiliation(s)
- Kwok-Leung Ong
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Liang Wu
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | | | - Rachel L O'Connell
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Hong Kong; State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Alicia J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Anthony C Keech
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Rudman-Melnick V, Adam M, Potter A, Chokshi SM, Ma Q, Drake KA, Schuh MP, Kofron JM, Devarajan P, Potter SS. Single-Cell Profiling of AKI in a Murine Model Reveals Novel Transcriptional Signatures, Profibrotic Phenotype, and Epithelial-to-Stromal Crosstalk. J Am Soc Nephrol 2020; 31:2793-2814. [PMID: 33115917 DOI: 10.1681/asn.2020010052] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Current management of AKI, a potentially fatal disorder that can also initiate or exacerbate CKD, is merely supportive. Therefore, deeper understanding of the molecular pathways perturbed in AKI is needed to identify targets with potential to lead to improved treatment. METHODS We performed single-cell RNA sequencing (scRNA-seq) with the clinically relevant unilateral ischemia-reperfusion murine model of AKI at days 1, 2, 4, 7, 11, and 14 after AKI onset. Using real-time quantitative PCR, immunofluorescence, Western blotting, and both chromogenic and single-molecule in situ hybridizations, we validated AKI signatures in multiple experiments. RESULTS Our findings show the time course of changing gene expression patterns for multiple AKI stages and all renal cell types. We observed elevated expression of crucial injury response factors-including kidney injury molecule-1 (Kim1), lipocalin 2 (Lcn2), and keratin 8 (Krt8)-and of several novel genes (Ahnak, Sh3bgrl3, and Col18a1) not previously examined in kidney pathologies. AKI induced proximal tubule dedifferentiation, with a pronounced nephrogenic signature represented by Sox4 and Cd24a. Moreover, AKI caused the formation of "mixed-identity cells" (expressing markers of different renal cell types) that are normally seen only during early kidney development. The injured tubules acquired a proinflammatory and profibrotic phenotype; moreover, AKI dramatically modified ligand-receptor crosstalk, with potential pathologic epithelial-to-stromal interactions. Advancing age in AKI onset was associated with maladaptive response and kidney fibrosis. CONCLUSIONS The scRNA-seq, comprehensive, cell-specific profiles provide a valuable resource for examining molecular pathways that are perturbed in AKI. The results fully define AKI-associated dedifferentiation programs, potential pathologic ligand-receptor crosstalk, novel genes, and the improved injury response in younger mice, and highlight potential targets of kidney injury.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Andrew Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Saagar M Chokshi
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Meredith P Schuh
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| |
Collapse
|
20
|
Lin X, Onda DA, Yang CH, Lewis JR, Levinger I, Loh K. Roles of bone-derived hormones in type 2 diabetes and cardiovascular pathophysiology. Mol Metab 2020; 40:101040. [PMID: 32544571 PMCID: PMC7348059 DOI: 10.1016/j.molmet.2020.101040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background Emerging evidence demonstrates that bone is an endocrine organ capable of influencing multiple physiological and pathological processes through the secretion of hormones. Recent research suggests complex crosstalk between the bone and other metabolic and cardiovascular tissues. It was uncovered that three of these bone-derived hormones—osteocalcin, lipocalin 2, and sclerostin—are involved in the endocrine regulations of cardiometabolic health and play vital roles in the pathophysiological process of developing cardiometabolic syndromes such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation is one of the hallmarks of cardiometabolic diseases and a major contributor to disease progression. Novel evidence also implicates important roles of bone-derived hormones in the regulation of chronic inflammation. Scope of review In this review, we provide a detailed overview of the physiological and pathological roles of osteocalcin, lipocalin 2, and sclerostin in cardiometabolic health regulation and disease development, with a focus on the modulation of chronic inflammation. Major conclusions Evidence supports that osteocalcin has a protective role in cardiometabolic health, and an increase of lipocalin 2 contributes to the development of cardiometabolic diseases partly via pro-inflammatory effects. The roles of sclerostin appear to be complicated: It exerts pro-adiposity and pro-insulin resistance effects in type 2 diabetes and has an anti-calcification effect during cardiovascular disease. A better understanding of the actions of these bone-derived hormones in the pathophysiology of cardiometabolic diseases will provide crucial insights to help further research develop new therapeutic strategies to treat these diseases.
Collapse
Affiliation(s)
- Xuzhu Lin
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia.
| | - Danise-Ann Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Joshua R Lewis
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Medical School, University of Western Australia, Perth, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
21
|
Bogorodskaya M, Fitch KV, Burdo TH, Maehler P, Easly RM, Murray GR, Feldpausch M, Adler GK, Grinspoon SK, Srinivasa S. Serum Lipocalin 2 (Neutrophil Gelatinase-Associated Lipocalin) in Relation to Biomarkers of Inflammation and Cardiac Stretch During Activation of the Renin-Angiotensin-Aldosterone System in Human Immunodeficiency Virus. J Infect Dis 2020; 220:1420-1424. [PMID: 31298286 DOI: 10.1093/infdis/jiz346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
PURPOSE To evaluate the relationship of lipocalin 2 to inflammation and cardiac injury with increased aldosterone in human immunodeficiency virus (HIV). METHODS A standardized 6-day low-sodium diet was used to stimulate renin-angiotensin-aldosterone system (RAAS) activation, and serum lipocalin 2 and biomarkers of inflammation and cardiac stretch were assessed among persons with or without HIV. RESULTS Lipocalin 2 levels increased with RAAS activation compared with suppression in the HIV group (median level [interquartile range], 71.3 [59.2-99.7] vs 67.0 [51.8-86.3] ng/mL; P = .01). During RAAS activation, lipocalin 2 was related to biomarkers of inflammation (tumor necrosis factor α [P = .007]), monocyte/macrophage activation (soluble CD163 [P = .005] and chemokine [C-C motif] ligand 2 [P = .03]), and markers of cardiac stretch (brain natriuretic peptide [P < .001] and N-terminal fragment of the prohormone brain natriuretic peptide [P = .001]) in HIV. CONCLUSION Lipocalin 2 may be important in modulating aldosterone-induced inflammation, monocyte activation, and cardiac stretch during RAAS activation in HIV. CLINICAL TRIAL REGISTRATION NCT01407237.
Collapse
Affiliation(s)
- Milana Bogorodskaya
- Division of Infectious Disease, Beth Israel Deaconess Medical Center and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Kathleen V Fitch
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Tricia H Burdo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Patrick Maehler
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Rebecca M Easly
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Gillian R Murray
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Meghan Feldpausch
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Gail K Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Steven K Grinspoon
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| | - Suman Srinivasa
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, Pennsylvania
| |
Collapse
|
22
|
Li D, Yan Sun W, Fu B, Xu A, Wang Y. Lipocalin-2-The myth of its expression and function. Basic Clin Pharmacol Toxicol 2019; 127:142-151. [PMID: 31597008 DOI: 10.1111/bcpt.13332] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/19/2019] [Indexed: 01/01/2023]
Abstract
Lipocalin-2 is a functional biomarker for acute and chronic kidney diseases, heart failure and obesity-related medical complications. It is rapidly induced in epithelial cells under stress conditions, but constitutively produced from pre-adipocytes and mature adipocytes. Measuring the lipocalin-2 levels represents an effective approach for risk prediction, patient stratification and disease management. Nevertheless, due to ligand-binding, post-translational modification and protein-protein interaction, lipocalin-2 exists as multiple variants that elicit different pathophysiological functions. To characterize the specific structure-functional relationships of lipocalin-2 variants is critical for the development of biomarker assays with sufficient precision and reliability. Moreover, identifying the pathological forms of lipocalin-2 will provide new therapeutic targets and treatment approaches for obesity-related complications.
Collapse
Affiliation(s)
- Dahui Li
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Wai Yan Sun
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Bowen Fu
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Chella Krishnan K, Sabir S, Shum M, Meng Y, Acín-Pérez R, Lang JM, Floyd RR, Vergnes L, Seldin MM, Fuqua BK, Jayasekera DW, Nand SK, Anum DC, Pan C, Stiles L, Péterfy M, Reue K, Liesa M, Lusis AJ. Sex-specific metabolic functions of adipose Lipocalin-2. Mol Metab 2019; 30:30-47. [PMID: 31767179 PMCID: PMC6812340 DOI: 10.1016/j.molmet.2019.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Lipocalin-2 (LCN2) is a secreted protein involved in innate immunity and has also been associated with several cardiometabolic traits in both mouse and human studies. However, the causal relationship of LCN2 to these traits is unclear, and most studies have examined only males. METHODS Using adeno-associated viral vectors we expressed LCN2 in either adipose or liver in a tissue specific manner on the background of a whole-body Lcn2 knockout or wildtype mice. Metabolic phenotypes including body weight, body composition, plasma and liver lipids, glucose homeostasis, insulin resistance, mitochondrial phenotyping, and metabolic cage studies were monitored. RESULTS We studied the genetics of LCN2 expression and associated clinical traits in both males and females in a panel of 100 inbred strains of mice (HMDP). The natural variation in Lcn2 expression across the HMDP exhibits high heritability, and genetic mapping suggests that it is regulated in part by Lipin1 gene variation. The correlation analyses revealed striking tissue dependent sex differences in obesity, insulin resistance, hepatic steatosis, and dyslipidemia. To understand the causal relationships, we examined the effects of expression of LCN2 selectively in liver or adipose. On a Lcn2-null background, LCN2 expression in white adipose promoted metabolic disturbances in females but not males. It acted in an autocrine/paracrine manner, resulting in mitochondrial dysfunction and an upregulation of inflammatory and fibrotic genes. On the other hand, on a null background, expression of LCN2 in liver had no discernible impact on the traits examined despite increasing the levels of circulating LCN2 more than adipose LCN2 expression. The mechanisms underlying the sex-specific action of LCN2 are unclear, but our results indicate that adipose LCN2 negatively regulates its receptor, LRP2 (or megalin), and its repressor, ERα, in a female-specific manner and that the effects of LCN2 on metabolic traits are mediated in part by LRP2. CONCLUSIONS Following up on our population-based studies, we demonstrate that LCN2 acts in a highly sex- and tissue-specific manner in mice. Our results have important implications for human studies, emphasizing the importance of sex and the tissue source of LCN2.
Collapse
Affiliation(s)
| | - Simon Sabir
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Michaël Shum
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Yonghong Meng
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Rebeca Acín-Pérez
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Jennifer M Lang
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Raquel R Floyd
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marcus M Seldin
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Brie K Fuqua
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Dulshan W Jayasekera
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Sereena K Nand
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Diana C Anum
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Miklós Péterfy
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marc Liesa
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Mancuso P, Bouchard B. The Impact of Aging on Adipose Function and Adipokine Synthesis. Front Endocrinol (Lausanne) 2019; 10:137. [PMID: 30915034 PMCID: PMC6421296 DOI: 10.3389/fendo.2019.00137] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/13/2019] [Indexed: 02/04/2023] Open
Abstract
During the last 40 years, there has been a world-wide increase in both the prevalence of obesity and an increase in the number of persons over the age of 60 due to a decline in deaths from infectious disease and the nutrition transition in low and middle income nations. While the increase in the elderly population indicates improvements in global public health, this population may experience a diminished quality of life due to the negative impacts of obesity on age-associated inflammation. Aging alters adipose tissue composition and function resulting in insulin resistance and ectopic lipid storage. A reduction in brown adipose tissue activity, declining sex hormones levels, and abdominal adipose tissue expansion occur with advancing years through the redistribution of lipids from the subcutaneous to the visceral fat compartment. These changes in adipose tissue function and distribution influence the secretion of adipose tissue derived hormones, or adipokines, that promote a chronic state of low-grade systemic inflammation. Ultimately, obesity accelerates aging by enhancing inflammation and increasing the risk of age-associated diseases. The focus of this review is the impact of aging on adipose tissue distribution and function and how these effects influence the elaboration of pro and anti-inflammatory adipokines.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Graduate Program in Immunology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Peter Mancuso
| | - Benjamin Bouchard
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|