1
|
van Baak MA, Mariman ECM. Physiology of Weight Regain after Weight Loss: Latest Insights. Curr Obes Rep 2025; 14:28. [PMID: 40163180 PMCID: PMC11958498 DOI: 10.1007/s13679-025-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE OF REVIEW This review summarizes the most recent research on the physiology of weight regain. It describes developments in areas that are currently being addressed and that may indicate promising directions for future research. RECENT FINDINGS Weight regain occurs independent of the way prior weight loss is achieved, i.e. by lifestyle, surgery or pharmacotherapy. Recent novel findings regarding weight regain belong to four areas. First, the immune obesity memory of which besides persistent immune cells promoting weight regain cells have been found that reduce weight regain. Second, the gut microbiome where autologous transplantation can limit weight regain. Third, the composition of the weight loss with the percentage of lost fat-free mass being inverse to the amount of regained weight independent of the weight loss procedure. Fourth, appetite control where after weight loss altered hypothalamic activity promoting hunger and weight regain persists, possibly mediated by altered neurotensin responses. In all four areas more conclusive evidence for their role in weight regain still needs to be obtained. Most studies on physiological mechanisms of weight regain are associative in nature and the number of intervention studies is very limited. To bring the field further, carefully designed intervention studies taking into account the dynamic character of weight loss and weight regain are needed.
Collapse
Affiliation(s)
- Marleen A van Baak
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and life Sciences+, Maastricht University, Maastricht, The Netherlands.
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, Maastricht, 6200MD, The Netherlands.
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and life Sciences+, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
2
|
Vanhie JJ, Orloff LE, Tate A, Goode C, Collao N, Pisanko A, Power KA, DE Lisio M. Obesity Promotes Marrow-Derived Myeloid Cell Accumulation While Exercise Reduces Proliferative Signaling in Colon Cancer. Med Sci Sports Exerc 2025; 57:317-326. [PMID: 39350427 DOI: 10.1249/mss.0000000000003572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
PURPOSE Obesity increases colon cancer risk that has been previously linked to marrow-derived myeloid cells. We previously demonstrated that exercise training (EX) prevents colon cancer initiation, potentially through reduced myelopoiesis. However, it remains unknown whether early myeloid cell accumulation and inflammation in the colon precedes carcinogenesis with high-fat diet (HFD)-induced obesity, and if EX can attenuate these effects. We hypothesized that obesity would promote colon carcinogenesis that was preceded by myeloid cell accumulation and inflammation that would be attenuated by EX. METHODS C57BL/6 mice were randomized to a HFD or control (CON) diet for 8 weeks. The HFD mice switched to CON diet and all mice were given intraperitoneal injections of azoxymethane (AOM) to induce colon cancer and randomized into EX or sedentary (SED) conditions. RESULTS HFD mice developed more aberrant crypt foci (ACF), a marker for early carcinogenesis, compared with CON ( P < 0.01), and EX developed fewer ACF compared with SED ( P < 0.0001). Marrow-derived ( P < 0.001) CD206 + macrophages were elevated in HFD compared with CON at study week 16 ( P < 0.01). Marrow-derived CD206 - macrophages ( P < 0.05) and marrow-derived ( P < 0.05) CD206 + macrophages were more abundant in HFD compared with CON at study week 42. EX did not alter colon immune cell populations. β-catenin protein was higher in HFD compared with CON at study week 42 ( P < 0.05), and STAT3 protein content was lower at study week 28 with EX compared with SED ( P < 0.05). CONCLUSIONS The results suggest that obesity promotes colon ACF formation, potentially through early inflammatory myeloid cell accumulation. Despite attenuating ACF, EX did not alter myeloid cell accumulation in the colon, suggesting that EX inhibits ACF formation through alternative mechanisms which may include reduced β-catenin and STAT3 signaling.
Collapse
Affiliation(s)
- James J Vanhie
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | - Lisa Ek Orloff
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | - Alice Tate
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | - Cole Goode
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | | | - Anastasia Pisanko
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, CANADA
| | | | | |
Collapse
|
3
|
Alharithi YJ, Phillips EA, Wilson TD, Couvillion SP, Nicora CD, Darakjian P, Rakshe S, Fei SS, Counts BR, Metz TO, Searles RP, Kumar S, Maloyan A. Metabolomic and transcriptomic remodeling of bone marrow myeloid cells in response to maternal obesity. Am J Physiol Endocrinol Metab 2025; 328:E254-E271. [PMID: 39792089 DOI: 10.1152/ajpendo.00333.2024] [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: 08/27/2024] [Revised: 09/24/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025]
Abstract
Maternal obesity puts the offspring at high risk of developing obesity and cardiometabolic diseases in adulthood. Here, we utilized a mouse model of maternal high-fat diet (HFD)-induced obesity that recapitulates metabolic perturbations seen in humans. We show increased adiposity in the offspring of HFD-fed mothers (Off-HFD) when compared with the offspring of regular diet-fed mothers (Off-RD). We have previously reported significant immune perturbations in the bone marrow of newly weaned Off-HFD. Here, we hypothesized that lipid metabolism is altered in the bone marrow of Off-HFD versus Off-RD. To test this hypothesis, we investigated the lipidomic profile of bone marrow cells collected from 3-week-old Off-RD and Off-HFD. Diacylglycerols (DAGs), triacylglycerols (TAGs), sphingolipids, and phospholipids were remarkably different between the groups, independent of fetal sex. Levels of cholesteryl esters were significantly decreased in Off-HFD, suggesting reduced delivery of cholesterol. These were accompanied by age-dependent progression of mitochondrial dysfunction in bone marrow cells. We subsequently isolated CD11b+ myeloid cells from 3-wk-old mice and conducted metabolomic, lipidomic, and transcriptomic analyses. The lipidomic profiles of myeloid cells were similar to those of bone marrow cells and included increases in DAGs and decreased TAGs. Transcriptomics revealed altered expression of genes related to immune pathways, including macrophage alternative activation, B-cell receptors, and transforming growth factor-β signaling. All told, this study revealed lipidomic, metabolomic, and gene expression abnormalities in bone marrow cells broadly, and in bone marrow myeloid cells particularly, in the newly weaned offspring of mothers with obesity, which might at least partially explain the progression of metabolic and cardiovascular diseases in their adulthood.NEW & NOTEWORTHY Our data revealed significant immunometabolic perturbations in the bone marrow and myeloid cells in the newly weaned offspring born to mothers with obesity. Adaptation to an adverse maternal intrauterine environment affects bone marrow metabolism at a very young age and might affect responses to immune challenges that appear later in life, for example, infections or cancer.
Collapse
Affiliation(s)
- Yem J Alharithi
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Elysse A Phillips
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Tim D Wilson
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Sneha P Couvillion
- Biological Sciences Division, Pacific Northwest National Laboratory (PNNL), Richland, Washington, United States
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory (PNNL), Richland, Washington, United States
| | - Priscila Darakjian
- Massively Parallel Sequencing Shared Resource, Oregon Health & Science University, Portland, Oregon, United States
| | - Shauna Rakshe
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Portland, Oregon, United States
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Suzanne S Fei
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Portland, Oregon, United States
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Brittany R Counts
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory (PNNL), Richland, Washington, United States
| | - Robert P Searles
- Massively Parallel Sequencing Shared Resource, Oregon Health & Science University, Portland, Oregon, United States
| | - Sushil Kumar
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Alina Maloyan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
4
|
Chen X, Zeng C, Chen X, Sun J, Jin F, Chen Z, Song J. Relationship between three body obesity indicators, WWI, BMI, WtHR, and periodontitis. Sci Rep 2025; 15:697. [PMID: 39753692 PMCID: PMC11699209 DOI: 10.1038/s41598-024-83963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
Many scholars have studied the relationship between obesity and periodontitis. The relationship between different obesity indicators and periodontitis seems to vary. In this study, we would like to explore the relationship between three commonly used obesity indicators, WWI (weight-adjusted-waist index), BMI (body mass index), WtHR (waist-to-height ratio), and periodontitis, and try to find the most clinically valuable indicators. The NHANES 2009-2014 provided the data for this study. 10,289, 10,600 and 10,290 participants were included to explore the relationship between WWI, BMI, WtHR and periodontitis respectively. Multiple logistic regression, smooth curve fitting, threshold effects analysis, and subgroup analysis were used to explore their relationships. WWI and periodontitis positively correlated in model III (OR = 1.35; 95% CI, 1.26-1.45, p < 0.001). This result persisted after the quartile for WWI was created. Furthermore, in smooth curve fitting, the relationship between WWI and periodontitis with an inflection point of 12.68 was observed. In the fully adjusted model, WtHR also showed a positive correlation with periodontitis (OR = 2.94; 95% CI, 1.57-5.50, p < 0.001). However, BMI did not express a significant association with periodontitis either as a continuous or categorical variable. This cross-sectional study exposed a positive correlation between WWI, WtHR and periodontitis and this positive correlation showed an age difference.
Collapse
Affiliation(s)
- XingJin Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - ChongWen Zeng
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - XianRun Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - JiangLing Sun
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - FuQian Jin
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Zhu Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China.
- School of Stomatology, Zunyi Medical University, Zunyi, China.
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550005, Guizhou, China.
| |
Collapse
|
5
|
Li Z, Ngu R, Naik AA, Trinh K, Paharkova V, Liao H, Liu Y, Zhuang C, Le D, Pei H, Asante I, Mittelman SD, Louie S. Adipocyte maturation impacts daunorubicin disposition and metabolism. Eur J Clin Invest 2024; 54:e14307. [PMID: 39254480 DOI: 10.1111/eci.14307] [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: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Acute lymphoblastic leukaemia (ALL) is the most common type of childhood leukaemia with effective chemotherapeutic treatment. However, obesity has been associated with higher ALL chemoresistance rates and lower event-free survival rates. The molecular mechanism of how obesity promotes chemotherapy resistance is not well delineated. OBJECTIVES This study evaluated the effect of adipocyte maturation on sequestration and metabolism of chemotherapeutic drug daunorubicin (DNR). METHODS Using targeted LC-MS/MS multi-analyte assay, DNR sequestration and metabolism were studied in human preadipocyte and adipocyte cell lines, where expressions of DNR-metabolizing enzymes aldo-keto reductases (AKR) and carbonyl reductases (CBR) were also evaluated. In addition, to identify the most DNR-metabolizing AKR/CBR isoforms, recombinant human AKR and CBR enzymes were subject to DNR metabolism. The results were further validated by AKR-, CBR-specific inhibitors. RESULTS This report shows that adipocyte maturation upregulates expressions of AKR and CBR enzymes (by 4- to 60- folds, p < .05), which is positively associated with enhanced sequestration and metabolism of DNR in adipocytes compared to preadipocytes (by ~30%, p < .05). In particular, adipocyte maturation upregulates AKR1C3 and CBR1, which are the predominate metabolic enzyme isoforms responsible for DNR biotransformation to its metabolites. CONCLUSION Fat is an expandable tissue that can sequester and detoxify DNR when stimulated by obesity, likely through the upregulation of DNR-metabolizing enzymes AKR1C3 and CBR1. Our data partially explains why obese ALL patients may be more likely to become chemoresistant towards DNR, and provides evidence for potential clinical investigation targeting obesity to reduce DNR chemoresistance.
Collapse
Affiliation(s)
- Zeyang Li
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Rachael Ngu
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Aditya Anil Naik
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Kaitlyn Trinh
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Vladislava Paharkova
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Hanyue Liao
- College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Yulu Liu
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Cindy Zhuang
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Danh Le
- Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Hua Pei
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Isaac Asante
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Stan Louie
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Chang X, Song J, Du X, Sun J, Chen X, Zhang J, Luo Y, Bai G. Association between cardiometabolic index (CMI) and periodontitis in US adults: analysis of NHANES data (2009-2014). BMC Oral Health 2024; 24:1346. [PMID: 39506722 PMCID: PMC11539322 DOI: 10.1186/s12903-024-05119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The cardiometabolic index (CMI) is a new predictor of obesity-related diseases, but its link to periodontitis is under-researched. This study aims to examine the potential association between the CMI and periodontitis. METHODS We conducted a cross-sectional study using the National Health and Nutrition Examination Survey (NHANES) database (2009-2014) to examine the potential association between CMI and periodontitis. The study utilized a weighted multivariate logistic model to assess the association between TG, HDL-C, TG/HDL-C, obesity indices (BMI, WC, WHtR, CMI), and periodontitis, employing the area under the receiver operating characteristic curves (ROC) to estimate areas under the curve (AUC). Furthermore, generalized smooth curve fitting was conducted to examine the relationship between CMI and periodontitis. Finally, the study incorporated subgroup analysis and interaction tests to examine consistency across different populations. RESULTS TG/HDL-C, WHtR, and CMI were positively associated with periodontitis in the fully adjusted classification model. It was observed that for each unit increase in CMI, there was a corresponding 17.8% increase in the prevalence of periodontitis [1.178 (1.049, 1.322) 0.00562] and 18.7% increase in the prevalence of moderate/severe [1.187 (1.057, 1.334) 0.00376] in the fully adjusted model. When CMI was used as a categorical variable, the adjusted OR for periodontitis prevalence increased significantly with increasing CMI after adjusting for all potential covariates (T3 vs. T1: OR, 1.28 [1.06, 1.55], p < 0.05). The ROC analysis indicated that a larger area under the curve was found in the CMI [0.554 (0.538-0.570)] than in the WC [0.544 (0.528-0.560)] and WHtR [0.544 (0.528-0.561)]. Nonetheless, the discrepancy observed did not reach statistical significance (all p > 0.05). Further generalized smooth curve fitting and threshold effect models indicated a positive linear correlation between CMI and periodontitis. Moreover, there is no interactive association between TG/HDL-C, WHtR, CMI, and periodontitis. CONCLUSIONS This cross-sectional study revealed a positive relationship between CMI and periodontitis. Further prospective studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Xingtao Chang
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China
- Department of Prosthodontics, Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, 550005, China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Xue Du
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China
- Department of Prosthodontics, Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, 550005, China
| | - Jiangling Sun
- Department of Endodontics, Guiyang Stomatological Hospital, Guiyang, China
| | - Xianrun Chen
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China
- Department of Prosthodontics, Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, 550005, China
| | - Jiqin Zhang
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China
- Department of Prosthodontics, Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, 550005, China
| | - Yi Luo
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China.
- Department of Prosthodontics, Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, 550005, China.
| | - Guohui Bai
- School of Stomatology, Zunyi Medical University, Dalian Road, Huichuan District, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
7
|
Phillips EA, Alharithi YJ, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to mothers with obesity. Int J Obes (Lond) 2024; 48:1542-1551. [PMID: 38937647 DOI: 10.1038/s41366-024-01563-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/OBJECTIVES Intrauterine metabolic reprogramming occurs in mothers with obesity during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to women with obesity. METHODS Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art flow cytometry, and targeted metabolomics approach coupled with a Seahorse metabolic analyzer. RESULTS We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also show a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we found changes in the immune complexity of bone marrow cells and identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers. Our data also revealed increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. CONCLUSIONS Altogether, we demonstrate that the offspring of mothers with obesity show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
Affiliation(s)
- Elysse A Phillips
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
- The University of California San Francisco, San Francisco, CA, USA
| | - Yem J Alharithi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Leena Kadam
- Department of OB/GYN, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Lisa M Coussens
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sushil Kumar
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Alina Maloyan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
- The University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Takaya J, Tanabe Y, Nomura N, Minami M, Onuma C, Yamagishi M, Kaneko K. Platelet and white blood cell counts correlate with leptin and body mass index in Japanese adolescents. Clin Pediatr Endocrinol 2024; 33:207-213. [PMID: 39359671 PMCID: PMC11442701 DOI: 10.1297/cpe.2024-0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/01/2024] [Indexed: 10/04/2024] Open
Abstract
Obesity is associated with mild chronic inflammation, frequently observed along with increased platelet and white blood cell (WBC) levels in adults. We aimed to clarify the relationship between peripheral blood cell count, body mass index standard deviation score (BMI-SDS), and adipocytokine levels in obese adolescents. Participants included 31 patients with obesity (age: 13.1 ± 3.1 yr) and 28 normal-weight controls (age: 13.3 ± 1.9 yr). Obesity was defined as a percentage of overweight ≥ 20%; patients with type 2 diabetes were excluded. As sex differences were observed in blood cell counts, the analysis was performed after adjusting for sex differences. The obese group has significantly higher WBC, red blood cell, and platelet counts, as well as high serum leptin levels and Homeostasis Model Assessment of insulin resistance (HOMA-IR) scores compared with those of the control group. In all participants, BMI-SDS significantly correlated with WBC and platelet counts. Platelet count correlated with serum leptin and glucose levels, whereas WBC count correlated with serum leptin, insulin, HOMA-IR, and glucose levels. Statistical analysis showed that serum leptin level significantly influenced the platelet count and HOMA-IR score affected WBC count. Increased platelet and WBC counts in adolescents with obesity may increase the risk of thrombosis.
Collapse
Affiliation(s)
- Junji Takaya
- Department of Pediatrics, Kawachi General Hospital, Osaka, Japan
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Yuko Tanabe
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Naohiro Nomura
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Miki Minami
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Chikusi Onuma
- Department of Pediatrics, Kawachi General Hospital, Osaka, Japan
| | | | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| |
Collapse
|
9
|
Yan J, Zhang P, Liu X, Pan C, Shi G, Ye P, Zou X, Li X, Zheng X, Liu Y, Yang H. Obesity modulates hematopoietic stem cell fate decision via IL-1β induced p38/MAPK signaling pathway. Stem Cell Res Ther 2024; 15:336. [PMID: 39343910 PMCID: PMC11441115 DOI: 10.1186/s13287-024-03915-w] [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: 05/03/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Obesity is accompanied by inflammation, which significantly affects the homeostasis of the immune microenvironment. Hematopoietic stem cells (HSCs), residing primarily in the bone marrow, play a vital role in maintaining and producing diverse mature blood cell lineages for the adult hematopoietic and immune systems. However, how HSCs development is affected by obese-promoting inflammation, and the mechanism by which HSC hematopoietic potency is affected by inflammatory signals originating from the obese-promoting changes on bone marrow niche remain unclear. This study elucidates the relationship between obesity-promoting inflammation and HSC fate determination. METHODS The obesity mice model was established by feeding C57BL/6J mice a high-fat diet (HFD) containing 60% kcal fat. After 6 weeks, HSCs were analyzed using flow cytometry and identified key inflammation cytokine. Transcriptome sequencing techniques were used to discern the distinct pathways in HSCs. Ultimately, confirming the biological mechanism of obesity-induced HSC fate changes via Anakinra blocking specific inflammatory signals. RESULTS Obesity caused by HFD changed the physical and biochemical properties of the bone marrow niche. In the HFD mice, the population of long-term HSCs in the bone marrow was decreased and facilitated HSCs differentiation towards the myeloid lineage. In addition, HFD increased expression of the inflammatory factor IL-1β in the bone marrow, and a significantly increased expression of IL-1r1 and active p38/MAPK signaling pathway were detected in the HSCs. Inhibition of IL-1β further normalized the expression of genes in p38/MAPK pathway and reversed HSC fate. CONCLUSIONS These findings have been demonstrated that the p38/MAPK signaling pathway in HSCs is activated by elevated levels of IL-1β within the HSC niche in obese models, thereby regulating HSC differentiation. It suggested a direct link between obesity-promoting inflammation and myeloid differentiation bias of HSCs in the HFD mice.
Collapse
Affiliation(s)
- Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- School College of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Chengwei Pan
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Guolin Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Penghui Ye
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xiaohang Zou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xiang Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xinmin Zheng
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yu Liu
- Department of Cardiology, Nanjing University Medical School Afliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China.
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, Shaanxi, China.
- Center of Special Environmental Biomechanics and Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| |
Collapse
|
10
|
Dzubanova M, Benova A, Ferencakova M, Coupeau R, Tencerova M. Nutrition and Bone Marrow Adiposity in Relation to Bone Health. Physiol Res 2024; 73:S107-S138. [PMID: 38752771 PMCID: PMC11412336 DOI: 10.33549/physiolres.935293] [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: 12/02/2023] [Accepted: 04/29/2024] [Indexed: 09/04/2024] Open
Abstract
Bone remodeling is energetically demanding process. Energy coming from nutrients present in the diet contributes to function of different cell type including osteoblasts, osteocytes and osteoclasts in bone marrow participating in bone homeostasis. With aging, obesity and osteoporosis the function of key building blocks, bone marrow stromal cells (BMSCs), changes towards higher accumulation of bone marrow adipose tissue (BMAT) and decreased bone mass, which is affected by diet and sex dimorphism. Men and women have unique nutritional needs based on physiological and hormonal changes across the life span. However, the exact molecular mechanisms behind these pathophysiological conditions in bone are not well-known. In this review, we focus on bone and BMAT physiology in men and women and how this approach has been taken by animal studies. Furthermore, we discuss the different diet interventions and impact on bone and BMAT in respect to sex differences. We also discuss the future perspective on precision nutrition with a consideration of sex-based differences which could bring better understanding of the diet intervention in bone health and weight management.
Collapse
Affiliation(s)
- M Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 4, Czech Republic.
| | | | | | | | | |
Collapse
|
11
|
Alharithi YJ, Phillips EA, Wilson TD, Couvillion SP, Nicora CD, Darakjian P, Rakshe S, Fei SS, Counts B, Metz TO, Searles R, Kumar S, Maloyan A. Metabolomic and transcriptomic remodeling of bone marrow myeloid cells in response to maternal obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608809. [PMID: 39229218 PMCID: PMC11370391 DOI: 10.1101/2024.08.20.608809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Maternal obesity puts the offspring at high risk of developing obesity and cardio-metabolic diseases in adulthood. Here, using a mouse model of maternal high-fat diet (HFD)-induced obesity, we show that whole body fat content of the offspring of HFD-fed mothers (Off-HFD) increases significantly from very early age when compared to the offspring regular diet-fed mothers (Off-RD). We have previously shown significant metabolic and immune perturbations in the bone marrow of newly-weaned offspring of obese mothers. Therefore, we hypothesized that lipid metabolism is altered in the bone marrow Off-HFD in newly-weaned offspring of obese mothers when compared to the Off-RD. To test this hypothesis, we investigated the lipidomic profile of bone marrow cells collected from three-week-old offspring of regular and high fat diet-fed mothers. Diacylgycerols (DAGs), triacylglycerols (TAGs), sphingolipids and phospholipids, including plasmalogen, and lysophospholipids were remarkably different between the groups, independent of fetal sex. Levels of cholesteryl esters were significantly decreased in offspring of obese mothers, suggesting reduced delivery of cholesterol to bone marrow cells. This was accompanied by age-dependent progression of mitochondrial dysfunction in bone marrow cells. We subsequently isolated CD11b+ myeloid cells from three-week-old mice and conducted metabolomics, lipidomics, and transcriptomics analyses. The lipidomic profiles of these bone marrow myeloid cells were largely similar to that seen in bone marrow cells and included increases in DAGs and phospholipids alongside decreased TAGs, except for long-chain TAGs, which were significantly increased. Our data also revealed significant sex-dependent changes in amino acids and metabolites related to energy metabolism. Transcriptomic analysis revealed altered expression of genes related to major immune pathways including macrophage alternative activation, B-cell receptor signaling, TGFβ signaling, and communication between the innate and adaptive immune systems. All told, this study revealed lipidomic, metabolomic, and gene expression abnormalities in bone marrow cells broadly, and in bone marrow myeloid cells particularly, in the newly-weaned offspring of obese mothers, which might at least partially explain the progression of metabolic and cardiovascular diseases in their adulthood.
Collapse
|
12
|
Dzubanova M, Bond JM, Craige SM, Tencerova M. NOX4-reactive oxygen species axis: critical regulators of bone health and metabolism. Front Cell Dev Biol 2024; 12:1432668. [PMID: 39188529 PMCID: PMC11345137 DOI: 10.3389/fcell.2024.1432668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Bone marrow stromal cells (BMSCs) play a significant role in bone metabolism as they can differentiate into osteoblasts, bone marrow adipocytes (BMAds), and chondrocytes. BMSCs chronically exposed to nutrient overload undergo adipogenic programming, resulting in bone marrow adipose tissue (BMAT) formation. BMAT is a fat depot transcriptionally, metabolically, and morphologically distinct from peripheral adipose depots. Reactive oxygen species (ROS) are elevated in obesity and serve as important signals directing BMSC fate. ROS produced by the NADPH oxidase (NOX) family of enzymes, such as NOX4, may be responsible for driving BMSC adipogenesis at the expense of osteogenic differentiation. The dual nature of ROS as both cellular signaling mediators and contributors to oxidative stress complicates their effects on bone metabolism. This review discusses the complex interplay between ROS and BMSC differentiation in the context of metabolic bone diseases.Special attention is paid to the role of NOX4-ROS in regulating cellular processes within the bone marrow microenvironment and potential target in metabolic bone diseases.
Collapse
Affiliation(s)
- Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Jacob M. Bond
- Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, VA, United States
| | - Siobhan M. Craige
- Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, United States
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Reytor-González C, Parise-Vasco JM, González N, Simancas-Racines A, Zambrano-Villacres R, Zambrano AK, Simancas-Racines D. Obesity and periodontitis: a comprehensive review of their interconnected pathophysiology and clinical implications. Front Nutr 2024; 11:1440216. [PMID: 39171112 PMCID: PMC11335523 DOI: 10.3389/fnut.2024.1440216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Obesity and periodontitis are significant health problems with a complex bidirectional relationship. Excess body fat is linked to systemic diseases and can lead to persistent inflammation, potentially harming periodontal health. Periodontitis, a chronic inflammatory condition affecting the supporting structures of teeth, poses substantial health risks. Both conditions share pathological processes such as inflammation and oxidative stress, which aggravate health status and make treatment more challenging. Understanding this interaction is crucial for developing effective management strategies for both diseases. This study explores the multifaceted aspects of obesity and periodontitis and their reciprocal relationship.
Collapse
Affiliation(s)
- Claudia Reytor-González
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| | - Juan Marcos Parise-Vasco
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| | - Natali González
- Facultad de Odontología, Universidad UTE, Santo Domingo, Ecuador
| | - Alison Simancas-Racines
- Carrera de Medicina Veterinaria, Facultad de Ciencias Agropecuarias y Recursos Naturales, Universidad Técnica de Cotopaxi, Latacunga, Ecuador
| | | | - Ana Karina Zambrano
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Daniel Simancas-Racines
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| |
Collapse
|
14
|
Balcaen T, Benova A, de Jong F, de Oliveira Silva R, Cajka T, Sakellariou D, Tencerova M, Kerckhofs G, De Borggraeve WM. Exploring contrast-enhancing staining agents for studying adipose tissue through contrast-enhanced computed tomography. J Lipid Res 2024; 65:100572. [PMID: 38823780 PMCID: PMC11259937 DOI: 10.1016/j.jlr.2024.100572] [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: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024] Open
Abstract
Contrast-enhanced computed tomography offers a nondestructive approach to studying adipose tissue in 3D. Several contrast-enhancing staining agents (CESAs) have been explored, whereof osmium tetroxide (OsO4) is the most popular nowadays. However, due to the toxicity and volatility of the conventional OsO4, alternative CESAs with similar staining properties were desired. Hf-WD 1:2 POM and Hexabrix have proven effective for structural analysis of adipocytes using contrast-enhanced computed tomography but fail to provide chemical information. This study introduces isotonic Lugol's iodine (IL) as an alternative CESA for adipose tissue analysis, comparing its staining potential with Hf-WD 1:2 POM and Hexabrix in murine caudal vertebrae and bovine muscle tissue strips. Single and sequential staining protocols were compared to assess the maximization of information extraction from each sample. The study investigated interactions, distribution, and reactivity of iodine species towards biomolecules using simplified model systems and assesses the potential of the CESA to provide chemical information. (Bio)chemical analyses on whole tissues revealed that differences in adipocyte gray values post-IL staining were associated with chemical distinctions between bovine muscle tissue and murine caudal vertebrae. More specific, a difference in the degree of unsaturation of fatty acids was identified as a likely contributor, though not the sole determinant of gray value differences. This research sheds light on the potential of IL as a CESA, offering both structural and chemical insights into adipose tissue composition.
Collapse
Affiliation(s)
- Tim Balcaen
- MolDesignS, Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Leuven, Belgium; Institute of Mechanics, Materials and Civil Engineering, Mechatronic, Electrical Energy and Dynamic Systems, UCLouvain, Louvain-la-Neuve, Belgium; Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Flip de Jong
- Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Rodrigo de Oliveira Silva
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Dimitrios Sakellariou
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular Systems, KU Leuven, Leuven, Belgium
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Greet Kerckhofs
- Institute of Mechanics, Materials and Civil Engineering, Mechatronic, Electrical Energy and Dynamic Systems, UCLouvain, Louvain-la-Neuve, Belgium; Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium; Department Materials Engineering, KU Leuven, Leuven, Belgium.
| | - Wim M De Borggraeve
- MolDesignS, Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Hasson M, Fernandes LM, Solomon H, Pepper T, Huffman NL, Pucha SA, Bariteau JT, Kaiser JM, Patel JM. Considering the Cellular Landscape in Marrow Stimulation Techniques for Cartilage Repair. Cells Tissues Organs 2024; 213:523-537. [PMID: 38599194 PMCID: PMC11633897 DOI: 10.1159/000538530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Marrow stimulation is a common reparative approach to treat injuries to cartilage and other soft tissues (e.g., rotator cuff). It involves the recruitment of bone marrow elements and mesenchymal stem cells (MSCs) into the defect, theoretically initiating a regenerative process. However, the resulting repair tissue is often weak and susceptible to deterioration with time. The populations of cells at the marrow stimulation site (beyond MSCs), and their contribution to inflammation, vascularity, and fibrosis, may play a role in quality of the repair tissue. SUMMARY In this review, we accomplish three goals: (1) systematically review clinical trials on the augmentation of marrow stimulation and evaluate their assumptions on the biological elements recruited; (2) detail the cellular populations in bone marrow and their impact on healing; and (3) highlight emerging technologies and approaches that could better guide these specific cell populations towards enhanced cartilage or soft tissue formation. KEY MESSAGES We found that most clinical trials do not account for cell heterogeneity, nor do they specify the regenerative element recruited, and those that do typically utilize descriptions such as "clots," "elements," and "blood." Furthermore, our review of bone marrow cell populations demonstrates a dramatically heterogenous cell population, including hematopoietic cells, immune cells, fibroblasts, macrophages, and only a small population of MSCs. Finally, the field has developed numerous innovative techniques to enhance the chondrogenic potential (and reduce the anti-regenerative impacts) of these various cell types. We hope this review will guide approaches that account for cellular heterogeneity and improve marrow stimulation techniques to treat chondral defects.
Collapse
Affiliation(s)
- Maddie Hasson
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Lorenzo M. Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Hanna Solomon
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Tristan Pepper
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas L. Huffman
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Saitheja A. Pucha
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Jason T. Bariteau
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jarred M. Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| | - Jay M. Patel
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Department of Veterans Affairs, Decatur, GA, USA
| |
Collapse
|
16
|
Tikhonova IV, Dyukina AR, Grinevich AA, Shaykhutdinova ER, Safronova VG. Changed regulation of granulocyte NADPH oxidase activity in the mouse model of obesity-induced type 2 diabetes mellitus. Free Radic Biol Med 2024; 216:33-45. [PMID: 38479632 DOI: 10.1016/j.freeradbiomed.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
NADPH oxidase is a target of hyperglycemia in type 2 diabetes mellitus (T2DM), which causes dysregulation of enzyme. Alterations in regulation of NADPH oxidase activity mediated receptor and non-receptor signaling in bone marrow granulocytes of mice with obesity-induced T2DM were studied. The animals fed high fat diet (516 kcal/100 g) for 16 weeks. NADPH oxidase-related generation of reactive species (RS) at normo- and hyperthermia was estimated using chemiluminescent analysis. The redox status of the cells was assessed by Redox Sensor Red CC-1. Baseline biochemical indicators in blood (glucose, cholesterol, HDL and LDL levels) were significant higher in T2DM mice versus controls. Using specific inhibitors, signaling mediated by formyl peptide receptors (FPRs) to NADPH oxidase was shown to involve PLC, PKC, cytochrome p450 in both control and T2DM groups and PLA2 in controls. In T2DM regulation of NADPH oxidase activity via mFpr1, a high-affinity receptors, occurred with a significant increase of the role of PKC isoforms and suppression of PLA2 participation. Significant differences between this regulation via mFpr2, low-affinity receptors, were not found. Non-receptor activation of NADPH oxidase with ionomycin (Ca2+ ionophore) or phorbol ester (direct activator of PKC isoforms) did not revealed differences in the kinetic parameters between groups at 37 °C and 40 °C. When these agents were used together (synergistic effect), lower sensitivity of cells to ionophore was observed in T2DM at both temperatures. Redox status in responses to opsonized zymosan was higher in T2DM mice at 37 °C and similar to control levels at 40 °C. ROC-analysis identified Tmax, RS production and effect of opsonized zymosan as the most significant predictors for discriminating between groups. It was concluded that Ca2+-dependent/PKC-mediated regulation of NADPH oxidase activity was altered in BM granulocytes from diabetic mice.
Collapse
Affiliation(s)
- Irina V Tikhonova
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3, Pushchino, 142290, Russia.
| | - Alsu R Dyukina
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3, Pushchino, 142290, Russia
| | - Andrei A Grinevich
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3, Pushchino, 142290, Russia
| | - Elvira R Shaykhutdinova
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, Prospect Nauki, 6, Pushchino, 142290, Russia
| | - Valentina G Safronova
- Institute of Cell Biophysics of Russian Academy of Sciences, Institutskaya st., 3, Pushchino, 142290, Russia
| |
Collapse
|
17
|
Stevenson M, Srivastava A, Nacher M, Hall C, Palaia T, Lee J, Zhao CL, Lau R, Ali MAE, Park CY, Schlamp F, Heffron SP, Fisher EA, Brathwaite C, Ragolia L. The Effect of Diet Composition on the Post-operative Outcomes of Roux-en-Y Gastric Bypass in Mice. Obes Surg 2024; 34:911-927. [PMID: 38191966 PMCID: PMC11926884 DOI: 10.1007/s11695-023-07052-w] [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: 10/31/2023] [Revised: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Roux-en-Y gastric bypass (RYGB) leads to the improvement of many obesity-associated conditions. The degree to which post-operative macronutrient composition contributes to metabolic improvement after RYGB is understudied. METHODS A mouse model of RYGB was used to examine the effects of diet on the post-operative outcomes of RYGB. Obese mice underwent either Sham or RYGB surgery and were administered either chow or HFD and then monitored for an additional 8 weeks. RESULTS After RYGB, reductions to body weight, fat mass, and lean mass were similar regardless of diet. RYGB and HFD were independently detrimental to bone mineral density and plasma vitamin D levels. Independent of surgery, HFD accelerated hematopoietic stem and progenitor cell proliferation and differentiation and exhibited greater myeloid lineage commitment. Independent of diet, systemic iron deficiency was present after RYGB. In both Sham and RYGB groups, HFD increased energy expenditure. RYGB increased fecal energy loss, and HFD after RYGB increased fecal lipid content. RYGB lowered fasting glucose and liver glycogen levels but HFD had an opposing effect. Indices of insulin sensitivity improved independent of diet. HFD impaired improvements to dyslipidemia, NAFLD, and fibrosis. CONCLUSION Post-operative diet plays a significant role in determining the degree to which RYGB reverses obesity-induced metabolic abnormalities such as hyperglycemia, dyslipidemia, and NAFLD. Diet composition may be targeted in order to assist in the treatment of post-RYGB bone mineral density loss and vitamin D deficiency as well as to reverse myeloid lineage commitment. HFD after RYGB continues to pose a significant multidimensional health risk.
Collapse
Affiliation(s)
- Matthew Stevenson
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Ankita Srivastava
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Maria Nacher
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Christopher Hall
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Thomas Palaia
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Jenny Lee
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Chaohui Lisa Zhao
- Department of Pathology, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Raymond Lau
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
- Department of Endocrinology, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Mohamed A E Ali
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Christopher Y Park
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Florencia Schlamp
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Sean P Heffron
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Collin Brathwaite
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
- Department of Surgery, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Louis Ragolia
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA.
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
18
|
Kumar V, Stewart JH. Obesity, bone marrow adiposity, and leukemia: Time to act. Obes Rev 2024; 25:e13674. [PMID: 38092420 DOI: 10.1111/obr.13674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 02/28/2024]
Abstract
Obesity has taken the face of a pandemic with less direct concern among the general population and scientific community. However, obesity is considered a low-grade systemic inflammation that impacts multiple organs. Chronic inflammation is also associated with different solid and blood cancers. In addition, emerging evidence demonstrates that individuals with obesity are at higher risk of developing blood cancers and have poorer clinical outcomes than individuals in a normal weight range. The bone marrow is critical for hematopoiesis, lymphopoiesis, and myelopoiesis. Therefore, it is vital to understand the mechanisms by which obesity-associated changes in BM adiposity impact leukemia development. BM adipocytes are critical to maintain homeostasis via different means, including immune regulation. However, obesity increases BM adiposity and creates a pro-inflammatory environment to upregulate clonal hematopoiesis and a leukemia-supportive environment. Obesity further alters lymphopoiesis and myelopoiesis via different mechanisms, which dysregulate myeloid and lymphoid immune cell functions mentioned in the text under different sequentially discussed sections. The altered immune cell function during obesity alters hematological malignancies and leukemia susceptibility. Therefore, obesity-induced altered BM adiposity, immune cell generation, and function impact an individual's predisposition and severity of leukemia, which should be considered a critical factor in leukemia patients.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John H Stewart
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
19
|
Moscatelli F, Monda A, Messina G, Picciocchi E, Monda M, Di Padova M, Monda V, Mezzogiorno A, Dipace A, Limone P, Messina A, Polito R. Exploring the Interplay between Bone Marrow Stem Cells and Obesity. Int J Mol Sci 2024; 25:2715. [PMID: 38473961 DOI: 10.3390/ijms25052715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Obesity, a complex disorder with rising global prevalence, is a chronic, inflammatory, and multifactorial disease and it is characterized by excessive adipose tissue accumulation and associated comorbidities. Adipose tissue (AT) is an extremely diverse organ. The composition, structure, and functionality of AT are significantly influenced by characteristics specific to everyone, in addition to the variability connected to various tissue types and its location-related heterogeneity. Recent investigation has shed light on the intricate relationship between bone marrow stem cells and obesity, revealing potential mechanisms that contribute to the development and consequences of this condition. Mesenchymal stem cells within the bone marrow, known for their multipotent differentiation capabilities, play a pivotal role in adipogenesis, the process of fat cell formation. In the context of obesity, alterations in the bone marrow microenvironment may influence the differentiation of mesenchymal stem cells towards adipocytes, impacting overall fat storage and metabolic balance. Moreover, bone marrow's role as a crucial component of the immune system adds another layer of complexity to the obesity-bone marrow interplay. This narrative review summarizes the current research findings on the connection between bone marrow stem cells and obesity, highlighting the multifaceted roles of bone marrow in adipogenesis and inflammation.
Collapse
Affiliation(s)
- Fiorenzo Moscatelli
- Department of Wellbeing, Nutrition and Sport, Pegaso Telematic University, 80143 Naples, Italy
| | - Antonietta Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Elisabetta Picciocchi
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Marilena Di Padova
- Department of Humanistic Studies, University of Foggia, 71100 Foggia, Italy
| | - Vincenzo Monda
- Department of Exercise Sciences and Well-Being, University of Naples "Parthenope", 80138 Naples, Italy
| | - Antonio Mezzogiorno
- Department of Mental Health, Fisics and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anna Dipace
- Department of Wellbeing, Nutrition and Sport, Pegaso Telematic University, 80143 Naples, Italy
| | - Pierpaolo Limone
- Department of Wellbeing, Nutrition and Sport, Pegaso Telematic University, 80143 Naples, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
20
|
Chen R, Armamento-Villareal R. Obesity and Skeletal Fragility. J Clin Endocrinol Metab 2024; 109:e466-e477. [PMID: 37440585 PMCID: PMC10795939 DOI: 10.1210/clinem/dgad415] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/15/2023]
Abstract
Skeletal fracture has recently emerged as a complication of obesity. Given the normal or better than normal bone mineral density (BMD), the skeletal fragility of these patients appears to be a problem of bone quality rather than quantity. Type 2 diabetes mellitus (T2DM), the incidence of which increases with increasing body mass index, is also associated with an increased risk for fractures despite a normal or high BMD. With the additional bone pathology from diabetes itself, patients with both obesity and T2DM could have a worse skeletal profile. Clinically, however, there are no available methods for identifying those who are at higher risk for fractures or preventing fractures in this subgroup of patients. Weight loss, which is the cornerstone in the management of obesity (with or without T2DM), is also associated with an increased risk of bone loss. This review of the literature will focus on the skeletal manifestations associated with obesity, its interrelationship with the bone defects associated with T2DM, and the available approach to the bone health of patients suffering from obesity.
Collapse
Affiliation(s)
- Rui Chen
- Division of Endocrinology, Diabetes and Metabolism at Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Reina Armamento-Villareal
- Division of Endocrinology, Diabetes and Metabolism at Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| |
Collapse
|
21
|
El-Masri BM, Leka B, Mustapha F, Gundesen MT, Hinge M, Lund T, Andersen TL, Diaz-delCastillo M, Jafari A. Bone marrow adipocytes provide early sign for progression from MGUS to multiple myeloma. Oncotarget 2024; 15:20-26. [PMID: 38227739 PMCID: PMC10791075 DOI: 10.18632/oncotarget.28548] [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: 10/13/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
Multiple Myeloma (MM) is the second most common hematological malignancy and is characterized by clonal expansion of malignant plasma cells in the bone marrow. In spite of recent advances in the field of MM, the disease has remained incurable. MM is preceded by a premalignant state known as monoclonal gammopathy of undetermined significance (MGUS), with a risk of progression to MM of 1% per year. Establishing a scalable approach that refines the identification of MGUS patients at high risk of progression to MM can transform the clinical management of the disease, improve the patient's quality of life, and will have significant socioeconomic implications. Here, we provide evidence that changes in the bone marrow adipose tissue (BMAT) provide an early sign for progression from MGUS to MM. We employed AI-assisted histological analysis of unstained bone marrow biopsies from MGUS subjects with or without progression to MM within 10 years (n = 24, n = 17 respectively). Although the BMAT fraction was not different between the two groups, bone marrow adipocyte (BMAd) density was decreased in MGUS patients who developed MM, compared to non-progressing MGUS patients. Importantly, the distribution profile for BMAd size and roundness was significantly different between the two groups, indicating a shift toward increased BMAd size and roundness in MGUS patients who developed MM. These early changes in the BMAT could serve as valuable early indicators for the transition from MGUS to MM, potentially enabling timely interventions and personalized treatment strategies. Finally, the AI-based approach for histological characterization of unstained bone marrow biopsies is cost-effective and fast, rendering its clinical implementation feasible.
Collapse
Affiliation(s)
- Bilal M. El-Masri
- Danish Spatial Imaging Consortium (DanSIC)
- Department of Clinical Research, Molecular Bone Histology (MBH) Lab, University of Southern Denmark, Odense, Denmark
| | - Benedeta Leka
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fatima Mustapha
- Department of Forensic Medicine, Molecular Bone Histology (MBH) Lab, University of Aarhus, Aarhus, Denmark
| | | | - Maja Hinge
- Department of Hematology, Lillebaelt Hospital, Vejle, Denmark
| | - Thomas Lund
- Department of Hematology Odense University Hospital, Odense, Denmark
| | - Thomas L. Andersen
- Danish Spatial Imaging Consortium (DanSIC)
- Department of Clinical Research, Molecular Bone Histology (MBH) Lab, University of Southern Denmark, Odense, Denmark
- Department of Forensic Medicine, Molecular Bone Histology (MBH) Lab, University of Aarhus, Aarhus, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Marta Diaz-delCastillo
- Danish Spatial Imaging Consortium (DanSIC)
- Department of Forensic Medicine, Molecular Bone Histology (MBH) Lab, University of Aarhus, Aarhus, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Alina M, Phillips E, Alharithi Y, Kadam L, Coussens L, Kumar S. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. RESEARCH SQUARE 2024:rs.3.rs-3830161. [PMID: 38313293 PMCID: PMC10836107 DOI: 10.21203/rs.3.rs-3830161/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to obese women. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also found a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
23
|
Govindarajah V, Sakabe M, Good S, Solomon M, Arasu A, Chen N, Zhang X, Grimes HL, Kendler A, Xin M, Reynaud D. Gestational diabetes in mice induces hematopoietic memory that affects the long-term health of the offspring. J Clin Invest 2024; 134:e169730. [PMID: 37988162 PMCID: PMC10786695 DOI: 10.1172/jci169730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023] Open
Abstract
Gestational diabetes is a common medical complication of pregnancy that is associated with adverse perinatal outcomes and an increased risk of metabolic diseases and atherosclerosis in adult offspring. The mechanisms responsible for this delayed pathological transmission remain unknown. In mouse models, we found that the development of atherosclerosis in adult offspring born to diabetic pregnancy can be in part linked to hematopoietic alterations. Although they do not show any gross metabolic disruptions, the adult offspring maintain hematopoietic features associated with diabetes, indicating the acquisition of a lasting diabetic hematopoietic memory. We show that the induction of this hematopoietic memory during gestation relies on the activity of the advanced glycation end product receptor (AGER) and the nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, which lead to increased placental inflammation. In adult offspring, we find that this memory is associated with DNA methyltransferase 1 (DNMT1) upregulation and epigenetic changes in hematopoietic progenitors. Together, our results demonstrate that the hematopoietic system can acquire a lasting memory of gestational diabetes and that this memory constitutes a pathway connecting gestational health to adult pathologies.
Collapse
Affiliation(s)
| | | | - Samantha Good
- Division of Experimental Hematology and Cancer Biology and
| | | | - Ashok Arasu
- Division of Experimental Hematology and Cancer Biology and
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology and
| | - Xuan Zhang
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - H. Leighton Grimes
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Pediatrics and
| | - Ady Kendler
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| |
Collapse
|
24
|
Pezzino S, Luca T, Castorina M, Puleo S, Latteri S, Castorina S. Role of Perturbated Hemostasis in MASLD and Its Correlation with Adipokines. Life (Basel) 2024; 14:93. [PMID: 38255708 PMCID: PMC10820028 DOI: 10.3390/life14010093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise, making it one of the most prevalent chronic liver disorders. MASLD encompasses a range of liver pathologies, from simple steatosis to metabolic dysfunction-associated steatohepatitis (MASH) with inflammation, hepatocyte damage, and fibrosis. Interestingly, the liver exhibits close intercommunication with fatty tissue. In fact, adipose tissue could contribute to the etiology and advancement of MASLD, acting as an endocrine organ that releases several hormones and cytokines, with the adipokines assuming a pivotal role. The levels of adipokines in the blood are altered in people with MASLD, and recent research has shed light on the crucial role played by adipokines in regulating energy expenditure, inflammation, and fibrosis in MASLD. However, MASLD disease is a multifaceted condition that affects various aspects of health beyond liver function, including its impact on hemostasis. The alterations in coagulation mechanisms and endothelial and platelet functions may play a role in the increased vulnerability and severity of MASLD. Therefore, more attention is being given to imbalanced adipokines as causative agents in causing disturbances in hemostasis in MASLD. Metabolic inflammation and hepatic injury are fundamental components of MASLD, and the interrelation between these biological components and the hemostasis pathway is delineated by reciprocal influences, as well as the induction of alterations. Adipokines have the potential to serve as the shared elements within this complex interrelationship. The objective of this review is to thoroughly examine the existing scientific knowledge on the impairment of hemostasis in MASLD and its connection with adipokines, with the aim of enhancing our comprehension of the disease.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
| | - Tonia Luca
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | | | - Stefano Puleo
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
| | - Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Sergio Castorina
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
25
|
Park E, Evans MA, Walsh K. Regulators of clonal hematopoiesis and physiological consequences of this condition. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:3. [PMID: 39119355 PMCID: PMC11309374 DOI: 10.20517/jca.2023.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Clonal hematopoiesis (CH) is a prevalent condition that results from somatic mutations in hematopoietic stem cells. When these mutations occur in "driver" genes, they can potentially confer fitness advantages to the affected cells, leading to a clonal expansion. While most clonal expansions of mutant cells are generally considered to be asymptomatic since they do not impact overall blood cell numbers, CH carriers face long-term risks of all-cause mortality and age-associated diseases, including cardiovascular disease and hematological malignancies. While considerable research has focused on understanding the association between CH and these diseases, less attention has been given to exploring the regulatory factors that contribute to the expansion of the driver gene clone. This review focuses on the association between environmental stressors and inherited genetic risk factors in the context of CH development. A better understanding of how these stressors impact CH development will facilitate mechanistic studies and potentially lead to new therapeutic avenues to treat individuals with this condition.
Collapse
Affiliation(s)
- Eunbee Park
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Kenneth Walsh
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
26
|
Phillips E, Alharithi Y, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569274. [PMID: 38077037 PMCID: PMC10705475 DOI: 10.1101/2023.11.29.569274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation, and reduction in levels of amino acids, a phenomenon previously linked to aging. Furthermore, in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, we identified a unique B cell population expressing CD19 and CD11b, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11b hi B cells, with all the populations being significantly more abundant in offspring of dams fed HFD but not a regular diet. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
27
|
Zhou HY, Feng X, Wang LW, Zhou R, Sun H, Chen X, Lu RB, Huang Y, Guo Q, Luo XH. Bone marrow immune cells respond to fluctuating nutritional stress to constrain weight regain. Cell Metab 2023; 35:1915-1930.e8. [PMID: 37703873 DOI: 10.1016/j.cmet.2023.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Weight regain after weight loss is a major challenge in the treatment of obesity. Immune cells adapt to fluctuating nutritional stress, but their roles in regulating weight regain remain unclear. Here, we identify a stem cell-like CD7+ monocyte subpopulation accumulating in the bone marrow (BM) of mice and humans that experienced dieting-induced weight loss. Adoptive transfer of CD7+ monocytes suppresses weight regain, whereas inducible depletion of CD7+ monocytes accelerates it. These cells, accumulating metabolic memories via epigenetic adaptations, preferentially migrate to the subcutaneous white adipose tissue (WAT), where they secrete fibrinogen-like protein 2 (FGL2) to activate the protein kinase A (PKA) signaling pathway and facilitate beige fat thermogenesis. Nevertheless, CD7+ monocytes gradually enter a quiescent state after weight loss, accompanied by increased susceptibility to weight regain. Notably, administration of FMS-like tyrosine kinase 3 ligand (FLT3L) remarkably rejuvenates CD7+ monocytes, thus ameliorating rapid weight regain. Together, our findings identify a unique bone marrow-derived metabolic-memory immune cell population that could be targeted to combat obesity.
Collapse
Affiliation(s)
- Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Li-Wen Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Heng Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ren-Bin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan 410008, China.
| |
Collapse
|
28
|
Diedrich JD, Cole CE, Pianko MJ, Colacino JA, Bernard JJ. Non-Toxicological Role of Aryl Hydrocarbon Receptor in Obesity-Associated Multiple Myeloma Cell Growth and Survival. Cancers (Basel) 2023; 15:5255. [PMID: 37958428 PMCID: PMC10649826 DOI: 10.3390/cancers15215255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity is not only a risk factor for multiple myeloma (MM) incidence, but it is also associated with an increased risk of progression from myeloma precursors-monoclonal gammopathy of undetermined significance-and smoldering myeloma. Adipocytes in the bone marrow (BMAs) microenvironment have been shown to facilitate MM cell growth via secreted factors, but the nature of these secreted factors and their mechanism of action have not been fully elucidated. The elevated expression of aryl hydrocarbon receptor (AhR) is associated with a variety of different cancers, including MM; however, the role of AhR activity in obesity-associated MM cell growth and survival has not been explored. Indeed, this is of particular interest as it has been recently shown that bone marrow adipocytes are a source of endogenous AhR ligands. Using multiple in vitro models of tumor-adipocyte crosstalk to mimic the bone microenvironment, we identified a novel, non-toxicological role of the adipocyte-secreted factors in the suppression of AhR activity in MM cells. A panel of six MM cell lines were cultured in the presence of bone marrow adipocytes in (1) a direct co-culture, (2) a transwell co-culture, or (3) an adipocyte-conditioned media to interrogate the effects of the secreted factors on MM cell AhR activity. Nuclear localization and the transcriptional activity of the AhR, as measured by CYP1A1 and CYP1B1 gene induction, were suppressed by exposure to BMA-derived factors. Additionally, decreased AhR target gene expression was associated with worse clinical outcomes. The knockdown of AhR resulted in reduced CYP1B1 expression and increased cellular growth. This tumor-suppressing role of CYP1A1 and CYP1B1 was supported by patient data which demonstrated an association between reduced target gene expression and worse overall survival. These data demonstrated a novel mechanism by which bone marrow adipocytes promote MM progression.
Collapse
Affiliation(s)
- Jonathan D. Diedrich
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Craig E. Cole
- Department of Medicine, Division of Hematology/Oncology, Michigan State University, East Lansing, MI 48910, USA;
- Karmanos Cancer Institute, McLaren Greater Lansing, Lansing, MI 48910, USA
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Matthew J. Pianko
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Justin A. Colacino
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
29
|
Sender R, Weiss Y, Navon Y, Milo I, Azulay N, Keren L, Fuchs S, Ben-Zvi D, Noor E, Milo R. The total mass, number, and distribution of immune cells in the human body. Proc Natl Acad Sci U S A 2023; 120:e2308511120. [PMID: 37871201 PMCID: PMC10623016 DOI: 10.1073/pnas.2308511120] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/11/2023] [Indexed: 10/25/2023] Open
Abstract
The immune system is a complex network of cells with critical functions in health and disease. However, a comprehensive census of the cells comprising the immune system is lacking. Here, we estimated the abundance of the primary immune cell types throughout all tissues in the human body. We conducted a literature survey and integrated data from multiplexed imaging and methylome-based deconvolution. We also considered cellular mass to determine the distribution of immune cells in terms of both number and total mass. Our results indicate that the immune system of a reference 73 kg man consists of 1.8 × 1012 cells (95% CI 1.5-2.3 × 1012), weighing 1.2 kg (95% CI 0.8-1.9). Lymphocytes constitute 40% of the total number of immune cells and 15% of the mass and are mainly located in the lymph nodes and spleen. Neutrophils account for similar proportions of both the number and total mass of immune cells, with most neutrophils residing in the bone marrow. Macrophages, present in most tissues, account for 10% of immune cells but contribute nearly 50% of the total cellular mass due to their large size. The quantification of immune cells within the human body presented here can serve to understand the immune function better and facilitate quantitative modeling of this vital system.
Collapse
Affiliation(s)
- Ron Sender
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yarden Weiss
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Navon
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Idan Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nofar Azulay
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Leeat Keren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shai Fuchs
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Elad Noor
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ron Milo
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
30
|
Benova A, Ferencakova M, Bardova K, Funda J, Prochazka J, Spoutil F, Cajka T, Dzubanova M, Balcaen T, Kerckhofs G, Willekens W, van Lenthe GH, Charyyeva A, Alquicer G, Pecinova A, Mracek T, Horakova O, Coupeau R, Hansen MS, Rossmeisl M, Kopecky J, Tencerova M. Omega-3 PUFAs prevent bone impairment and bone marrow adiposity in mouse model of obesity. Commun Biol 2023; 6:1043. [PMID: 37833362 PMCID: PMC10575870 DOI: 10.1038/s42003-023-05407-8] [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: 04/06/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Obesity adversely affects bone and fat metabolism in mice and humans. Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) have been shown to improve glucose metabolism and bone homeostasis in obesity. However, the impact of omega-3 PUFAs on bone marrow adipose tissue (BMAT) and bone marrow stromal cell (BMSC) metabolism has not been intensively studied yet. In the present study we demonstrated that omega-3 PUFA supplementation in high fat diet (HFD + F) improved bone parameters, mechanical properties along with decreased BMAT in obese mice when compared to the HFD group. Primary BMSCs isolated from HFD + F mice showed decreased adipocyte and higher osteoblast differentiation with lower senescent phenotype along with decreased osteoclast formation suggesting improved bone marrow microenvironment promoting bone formation in mice. Thus, our study highlights the beneficial effects of omega-3 PUFA-enriched diet on bone and cellular metabolism and its potential use in the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Michaela Ferencakova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Funda
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Tim Balcaen
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | - Arzuv Charyyeva
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Glenda Alquicer
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Pecinova
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Mracek
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Roman Coupeau
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Morten Svarer Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense, C DK-5000, Denmark
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
31
|
Leyderman M, Wilmore JR, Shope T, Cooney RN, Urao N. Impact of intestinal microenvironments in obesity and bariatric surgery on shaping macrophages. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00033. [PMID: 38037591 PMCID: PMC10683977 DOI: 10.1097/in9.0000000000000033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Obesity is associated with alterations in tissue composition, systemic cellular metabolism, and low-grade chronic inflammation. Macrophages are heterogenous innate immune cells ubiquitously localized throughout the body and are key components of tissue homeostasis, inflammation, wound healing, and various disease states. Macrophages are highly plastic and can switch their phenotypic polarization and change function in response to their local environments. Here, we discuss how obesity alters the intestinal microenvironment and potential key factors that can influence intestinal macrophages as well as macrophages in other organs, including adipose tissue and hematopoietic organs. As bariatric surgery can induce metabolic adaptation systemically, we discuss the potential mechanisms through which bariatric surgery reshapes macrophages in obesity.
Collapse
Affiliation(s)
- Michael Leyderman
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Joel R. Wilmore
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Timothy Shope
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Robert N. Cooney
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
32
|
Du Y, Gupta P, Qin S, Sieber M. The role of metabolism in cellular quiescence. J Cell Sci 2023; 136:jcs260787. [PMID: 37589342 PMCID: PMC10445740 DOI: 10.1242/jcs.260787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Cellular quiescence is a dormant, non-dividing cell state characterized by significant shifts in physiology and metabolism. Quiescence plays essential roles in a wide variety of biological processes, ranging from microbial sporulation to human reproduction and wound repair. Moreover, when the regulation of quiescence is disrupted, it can drive cancer growth and compromise tissue regeneration after injury. In this Review, we examine the dynamic changes in metabolism that drive and support dormant and transiently quiescent cells, including spores, oocytes and adult stem cells. We begin by defining quiescent cells and discussing their roles in key biological processes. We then examine metabolic factors that influence cellular quiescence in both healthy and disease contexts, and how these could be leveraged in the treatment of cancer.
Collapse
Affiliation(s)
- Yipeng Du
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, MC9040 ND13.214, Dallas, TX 75390, USA
| | - Parul Gupta
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, MC9040 ND13.214, Dallas, TX 75390, USA
| | - Shenlu Qin
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, MC9040 ND13.214, Dallas, TX 75390, USA
| | - Matthew Sieber
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, MC9040 ND13.214, Dallas, TX 75390, USA
| |
Collapse
|
33
|
AlShomar A, Abdulmonem WA, Ahmad QS, Alharbi MS, Alkhiari R, Hamad EM, Rasheed Z. Assessment of osteoporosis in patients with type 2 diabetes mellitus: A study from the central region of Saudi Arabia. Saudi Med J 2023; 44:711-716. [PMID: 37463708 PMCID: PMC10370382 DOI: 10.15537/smj.2023.44.7.20230238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES To understand the impact of diabetes on bone mineral density and whether it increases the likelihood of osteoporosis. METHODS This study was performed on 327 Saudis (aged >40 years) who were screened for osteoporosis and diabetes mellitus (DM). The levels of osteoporosis were determined by an estimation of Bone mineral density (BMD) using a DEXA scan examination. The data on BMD from diabetic subjects were compared with healthy nondiabetic controls. RESULTS Out of 327 enrolled subjects, 38 (11.6%) were found to be osteoporotic, whereas 138 (42.2%) had DM. The data showed that the number of patients with osteoporosis in the DM group was 14 (36.8%), significantly lower than in nondiabetic patients, 21 (55.2%) (p=0.0015). Notably, the data showed no significant difference in the mean BMD of the femur in patients with DM (0.926 g/cm2) and non-diabetes (0.936 g/cm2) (p=0.280; T-score p=0.4746). The mean BMD levels in the spine of the DM study group (1.049 g/cm2) were significantly higher when compared with nondiabetic healthy controls (0.990 g/cm2) (p=0.0031). CONCLUSION Patients with diabetes had higher lumbar BMD than nondiabetics, although femoral BMD was similar. Patients with diabetes have a lower osteoporosis risk than nondiabetics.
Collapse
Affiliation(s)
- Ahmad AlShomar
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Waleed Al Abdulmonem
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Qazi Shoeb Ahmad
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Mariam S. Alharbi
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Resheed Alkhiari
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Essam M. Hamad
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| | - Zafar Rasheed
- From the Department of Medicine (AlShomar, Alharbi, Alkhiari), College of Medicine; from the Department of Pathology (Al Abdulmonem, Rasheed), College of Medicine; from the Department of Statistics (Ahmad), Deanship of Educational Services; Department of Food Science and Human Nutrition (Hamad), College of Agriculture and Veterinary Medicine, Qassim University, Buraidah; and from Dr. Sulaiman Al Habib Medical Group (AlShomar, Alkhiari), Qassim, Kingdom of Saudi Arabia.
| |
Collapse
|
34
|
Yun N, Nah J, Lee MN, Wu D, Pae M. Post-Effects of Time-Restricted Feeding against Adipose Tissue Inflammation and Insulin Resistance in Obese Mice. Nutrients 2023; 15:2617. [PMID: 37299580 PMCID: PMC10255447 DOI: 10.3390/nu15112617] [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: 04/30/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Time-restricted feeding (TRF) has been shown to improve the disordered metabolic and immunologic functions associated with obesity, however little is known about its post-effects after the cessation of TRF practice. In the current study, we determined how long the effects of TRF persist, and whether the effects are tissue-dependent. There were four groups of mice in this study: overweight and obese mice were randomized into (1) TRF group (TRF for 6 weeks), (2) post-TRF group (TRF for 4 weeks and later ad libitum), (3) continuous ad libitum of high-fat diet (HFD-AL), and (4) the lean control-fed low-fat diet ad libitum. Blood, liver, and adipose tissues were collected to measure the metabolic, inflammatory, and immune cell parameters. The results showed that TRF withdrawal quickly led to increased body weight/adiposity and reversed fasting blood glucose. However, fasting insulin and insulin resistance index HOMA-IR remained lower in the post-TRF than in the HFD-AL group. In addition, TRF-induced reduction in blood monocytes waned in the post-TRF group, but the TRF effects on mRNA levels of proinflammatory immune cells (macrophages Adgre1 and Itgax) and cytokine (Tnf) in adipose tissue remained lower in the post-TRF group than in the HFD-AL group. Furthermore, the TRF group was protected from the down-regulation of Pparg mRNA expression in adipose tissue, which was also observed in the post-TRF group to a lesser extent. The post-TRF animals displayed liver mass similar to those in the TRF group, but the TRF effects on the mRNA of inflammation markers in the liver vanished completely. Together, these results indicate that, although the lasting effects of TRF may differ by tissues and genes, the impact of TRF on adipose tissue inflammation and immune cell infiltration could last a couple of weeks, which may, in part, contribute to the maintenance of insulin sensitivity even after the cessation of TRF.
Collapse
Affiliation(s)
- Narae Yun
- Department of Food and Nutrition, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea; (N.Y.); (J.N.)
| | - Jiyeon Nah
- Department of Food and Nutrition, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea; (N.Y.); (J.N.)
| | - Mi Nam Lee
- Department of Biological Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea;
| | - Dayong Wu
- Nutritional Immunology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, MA 02111, USA;
| | - Munkyong Pae
- Department of Food and Nutrition, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea; (N.Y.); (J.N.)
| |
Collapse
|
35
|
Kelem A, Shiferaw E, Adane T. Hematological abnormalities and associated factors among metabolic syndrome patients at the University of Gondar comprehensive specialized hospital, Northwest Ethiopia. PLoS One 2023; 18:e0286163. [PMID: 37228109 DOI: 10.1371/journal.pone.0286163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Metabolic Syndrome (MetS) is a cluster of interconnected metabolic diseases. Hematological abnormalities are common but neglected complications of MetS. Thus, this study aimed to determine the magnitude of hematological abnormalities and their associated factors among MetS patients at the University of Gondar comprehensive specialized hospital, Northwest Ethiopia. METHOD A hospital-based cross-sectional study was conducted at the University of Gondar comprehensive specialized hospital from March to May 2022. A total of 384 MetS patients were selected using a systematic random sampling technique. Data were collected using pre-tested structured questionnaires and checklists. Anthropometric and blood pressure measurements were taken, and blood sample was collected for complete blood count determination. Stool and blood film examinations were performed to detect intestinal and malaria parasites, respectively. Data were entered into EpiData 3.1 and analyzed by Stata 14.0 software. Bivariate and multivariate logistic regression models were fitted to identify factors associated with hematological abnormalities. A p-value of < 0.05 was considered statistically significant. RESULTS The magnitude of anemia, leukopenia, leukocytosis, thrombocytopenia, and thrombocytosis was found to be 13.3%, 0.5%, 2.9%, 1.6%, and 2.3%, respectively. Being male (AOR = 2.65, 95% CI: 1.14, 6.20), rural residency (AOR = 5.79, 95% CI: 1.72, 19.51), taking antihypertensive medications (AOR = 3.85, 95% CI: 1.16, 12.78), having elevated triglyceride level (AOR = 2.21, 95% CI: 1.03, 4.75), and being overweight or obese (AOR = 0.32, 95% CI: 0.16, 0.64) were significantly associated with anemia. CONCLUSIONS Anemia was the most prevalent hematological abnormality identified in the present study, followed by leukocytosis and thrombocytosis. Anemia was a mild public health problem among MetS patients in the study area. Routine anemia screening for all MetS patients, especially for those with significant associated factors, may help in the early detection and effective management of anemia, which subsequently improves the patients' quality of life.
Collapse
Affiliation(s)
- Amanuel Kelem
- Department of Medical Laboratory Sciences, Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Elias Shiferaw
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tiruneh Adane
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
36
|
Al-Azab M, Idiiatullina E, Safi M, Hezam K. Enhancers of mesenchymal stem cell stemness and therapeutic potency. Biomed Pharmacother 2023; 162:114356. [PMID: 37040673 DOI: 10.1016/j.biopha.2023.114356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 04/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can differentiate into a range of cell types, including osteoblasts, chondrocytes, myocytes, and adipocytes. Multiple preclinical investigations and clinical trials employed enhanced MSCs-dependent therapies in treatment of inflammatory and degenerative diseases. They have demonstrated considerable and prospective therapeutic potentials even though the large-scale use remains a problem. Several strategies have been used to improve the therapeutic potency of MSCs in cellular therapy. Treatment of MSCs utilizing pharmaceutical compounds, cytokines, growth factors, hormones, and vitamins have shown potential outcomes in boosting MSCs' stemness. In this study, we reviewed the current advances in enhancing techniques that attempt to promote MSCs' therapeutic effectiveness in cellular therapy and stemness in vivo with potential mechanisms and applications.
Collapse
Affiliation(s)
- Mahmoud Al-Azab
- Department of Immunology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Elina Idiiatullina
- Department of Immunology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China; Department of Therapy and Nursing, Bashkir State Medical University, Ufa 450008, Russia
| | - Mohammed Safi
- Department of Respiratory Diseases, Shandong Second Provincial General Hospital, Shandong University, Shandong, China
| | - Kamal Hezam
- Nankai University School of Medicine, Tianjin 300071, China; Department of Microbiology, Faculty of Applied Science, Taiz University, 6350 Taiz, Yemen
| |
Collapse
|
37
|
Marques-Mourlet C, Di Iorio R, Fairfield H, Reagan MR. Obesity and myeloma: Clinical and mechanistic contributions to disease progression. Front Endocrinol (Lausanne) 2023; 14:1118691. [PMID: 36909335 PMCID: PMC9996186 DOI: 10.3389/fendo.2023.1118691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Obesity and obesogenic behaviors are positively associated with both monoclonal gammopathy of unknown significance (MGUS) and multiple myeloma (MM). As the only known modifiable risk factor, this association has emerged as a new potential target for MM prevention, but little is known about the mechanistic relationship of body weight with MM progression. Here we summarize epidemiological correlations between weight, body composition, and the various stages of myeloma disease progression and treatments, as well as the current understanding of the molecular contributions of obesity-induced changes in myeloma cell phenotype and signaling. Finally, we outline groundwork for the future characterization of the relationship between body weight patterns, the bone marrow microenvironment, and MM pathogenesis in animal models, which have the potential to impact our understanding of disease pathogenesis and inform MM prevention messages.
Collapse
Affiliation(s)
- Constance Marques-Mourlet
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Strasbourg, Pharmacology Department, Strasbourg, France
| | - Reagan Di Iorio
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of New England, College of Osteopathic Medicine, Biddeford, ME, United States
| | - Heather Fairfield
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| |
Collapse
|
38
|
Fairfield H, Condruti R, Farrell M, Di Iorio R, Gartner CA, Vary C, Reagan MR. Development and characterization of three cell culture systems to investigate the relationship between primary bone marrow adipocytes and myeloma cells. Front Oncol 2023; 12:912834. [PMID: 36713534 PMCID: PMC9874147 DOI: 10.3389/fonc.2022.912834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 11/21/2022] [Indexed: 01/12/2023] Open
Abstract
The unique properties of the bone marrow (BM) allow for migration and proliferation of multiple myeloma (MM) cells while also providing the perfect environment for development of quiescent, drug-resistant MM cell clones. BM adipocytes (BMAds) have recently been identified as important contributors to systemic adipokine levels, bone strength, hematopoiesis, and progression of metastatic and primary BM cancers, such as MM. Recent studies in myeloma suggest that BMAds can be reprogrammed by tumor cells to contribute to myeloma-induced bone disease, and, reciprocally, BMAds support MM cells in vitro. Importantly, most data investigating BMAds have been generated using adipocytes generated by differentiating BM-derived mesenchymal stromal cells (BMSCs) into adipocytes in vitro using adipogenic media, due to the extreme technical challenges associated with isolating and culturing primary adipocytes. However, if studies could be performed with primary adipocytes, then they likely will recapitulate in vivo biology better than BMSC-derived adipocytes, as the differentiation process is artificial and differs from in vivo differentiation, and progenitor cell(s) of the primary BMAd (pBMAds) may not be the same as the BMSCs precursors used for adipogenic differentiation in vitro. Therefore, we developed and refined three methods for culturing pBMAds: two-dimensional (2D) coverslips, 2D transwells, and three-dimensional (3D) silk scaffolds, all of which can be cultured alone or with MM cells to investigate bidirectional tumor-host signaling. To develop an in vitro model with a tissue-like structure to mimic the BM microenvironment, we developed the first 3D, tissue engineered model utilizing pBMAds derived from human BM. We found that pBMAds, which are extremely fragile, can be isolated and stably cultured in 2D for 10 days and in 3D for up to 4 week in vitro. To investigate the relationship between pBMAds and myeloma, MM cells can be added to investigate physical relationships through confocal imaging and soluble signaling molecules via mass spectrometry. In summary, we developed three in vitro cell culture systems to study pBMAds and myeloma cells, which could be adapted to investigate many diseases and biological processes involving the BM, including other bone-homing tumor types.
Collapse
Affiliation(s)
- Heather Fairfield
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | | | - Mariah Farrell
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Reagan Di Iorio
- MaineHealth Institute for Research, Scarborough, ME, United States,University of New England, Biddeford, ME, United States
| | - Carlos A. Gartner
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Calvin Vary
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States,*Correspondence: Michaela R. Reagan,
| |
Collapse
|
39
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
40
|
Kim Y, Lee Y, Lee MN, Nah J, Yun N, Wu D, Pae M. Time-restricted feeding reduces monocyte production by controlling hematopoietic stem and progenitor cells in the bone marrow during obesity. Front Immunol 2022; 13:1054875. [PMID: 36569870 PMCID: PMC9771705 DOI: 10.3389/fimmu.2022.1054875] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Time-restricted feeding (TRF) has emerged as a promising dietary approach in improving metabolic parameters associated with obesity, but its effect on immune cells under obesogenic condition is poorly understood. We conducted this study to determine whether TRF exerts its therapeutic benefit over obesity-induced myeloid cell production by analyzing hematopoietic stem and progenitor cells in bone marrow (BM) and immune cell profile in circulation. Male C57BL/6 mice were fed a low-fat diet (LFD) or high-fat diet (HFD) ad libitum for 6 weeks and later a subgroup of HFD mice was switched to a daily 10 h-TRF schedule for another 6 weeks. Mice on HFD ad libitum for 12 weeks had prominent monocytosis and neutrophilia, associated with expansion of BM myeloid progenitors, such as multipotent progenitors, pre-granulocyte/macrophage progenitors, and granulocyte/macrophage progenitors. TRF intervention in overweight and obese mice diminished these changes to a level similar to those seen in mice fed LFD. While having no effect on BM progenitor cell proliferation, TRF reduced expression of Cebpa, a transcription factor required for myeloid differentiation. These results indicate that TRF intervention may help maintain immune cell homeostasis in BM and circulation during obesity, which may in part contribute to health benefits associated with TRF.
Collapse
Affiliation(s)
- Yelim Kim
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Youngyoon Lee
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Mi Nam Lee
- Department of Biological Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Jiyeon Nah
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Narae Yun
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Dayong Wu
- Nutritional Immunology Laboratory, Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Munkyong Pae
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea,*Correspondence: Munkyong Pae,
| |
Collapse
|
41
|
Vanhie JJ, Kim W, Ek Orloff L, Ngu M, Collao N, De Lisio M. The role of exercise-and high fat diet-induced bone marrow extracellular vesicles in stress hematopoiesis. Front Physiol 2022; 13:1054463. [PMID: 36505084 PMCID: PMC9728614 DOI: 10.3389/fphys.2022.1054463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Exercise and obesity regulate hematopoiesis, in part through alterations in cellular and soluble components of the bone marrow niche. Extracellular vesicles (EVs) are components of the bone marrow niche that regulate hematopoiesis; however, the role of exercise training or obesity induced EVs in regulating hematopoiesis remains unknown. To address this gap, donor EVs were isolated from control diet-fed, sedentary mice (CON-SED), control diet-fed exercise trained mice (CON-EX), high fat diet-fed, sedentary mice (HFD-SED), and high fat diet-fed, exercise trained mice (HFD-EX) and injected into recipient mice undergoing stress hematopoiesis. Hematopoietic and niche cell populations were quantified, and EV miRNA cargo was evaluated. EV content did not differ between the four groups. Mice receiving HFD-EX EVs had fewer hematopoietic stem cells (HSCs) (p < 0.01), long-term HSC (p < 0.05), multipotent progenitors (p < 0.01), common myeloid progenitors (p<0.01), common lymphoid progenitors (p < 0.01), and granulocyte-macrophage progenitors (p < 0.05), compared to mice receiving HFD-SED EVs. Similarly, mice receiving EX EVs had fewer osteoprogenitor cells compared to SED (p < 0.05) but enhanced mesenchymal stromal cell (MSC) osteogenic differentiation in vitro (p < 0.05) compared to SED EVs. HFD EVs enhanced mesenchymal stromal cell (MSC) adipogenesis in vitro (p < 0.01) compared to CON EVs. HFD-EX EVs had lower microRNA-193 and microRNA-331-5p content, microRNAs implicated in inhibiting osteogenesis and leukemic cell expansion respectively, compared to HFD-SED EVs. The results identify alterations in EV cargo as a novel mechanism by which exercise training alters stress hematopoiesis and the bone marrow niche.
Collapse
Affiliation(s)
- James J. Vanhie
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Wooseok Kim
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Lisa Ek Orloff
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Matthew Ngu
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Nicolas Collao
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,*Correspondence: Michael De Lisio,
| |
Collapse
|
42
|
The relationship between anemia and obesity. Expert Rev Hematol 2022; 15:911-926. [PMID: 36189499 DOI: 10.1080/17474086.2022.2131521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Obesity is linked to a variety of unfavourable outcomes, including anaemia, which is a serious global public health problem. The prevalence of obesity along with anaemia suggests a relationship between obesity and anaemia. Recent studies have demonstrated strong associations between anaemia and obesity, chronic diseases, ageing, hepato-renal impairment, chronic infection, autoimmune diseases, and widespread malignancy. Thus, the intersection point of obesity and anaemia is an important area of attention. AREA COVERED This paper reviews the pathophysiology of obesity and anaemia. Then, It deliberates the relationship between obesity and different types of anaemia and other clinical forms associated with anaemia. EXPERT OPINION Obesity, especially obesity-related to excessive visceral fat distribution, is accompanied by several disturbances at the endothelial, hormonal, and inflammatory levels. These disturbances induce activation of several mechanisms that contribute to the anaemic state. Over-weight patients with chronic anaemias are required to maintain the related vitamins and minerals at optimum levels and appropriate BMI. In addition, a regular clinical follow-up is essential to be scheduled to reduce the risk of complications associated with anaemia in obese patients.
Collapse
|
43
|
Interplay between fat cells and immune cells in bone: Impact on malignant progression and therapeutic response. Pharmacol Ther 2022; 238:108274. [DOI: 10.1016/j.pharmthera.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
|
44
|
Hellmich C, Wojtowicz EE. You are what you eat: How to best fuel your immune system. Front Immunol 2022; 13:1003006. [PMID: 36211413 PMCID: PMC9533172 DOI: 10.3389/fimmu.2022.1003006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 11/26/2022] Open
Abstract
Normal bone marrow (BM) homeostasis ensures consistent production of progenitor cells and mature blood cells. This requires a reliable supply of nutrients in particular free fatty acids, carbohydrates and protein. Furthermore, rapid changes can occur in response to stress such as infection which can alter the demand for each of these metabolites. In response to infection the haematopoietic stem cells (HSCs) must respond and expand rapidly to facilitate the process of emergency granulopoiesis required for the immediate immune response. This involves a shift from the use of glycolysis to oxidative phosphorylation for energy production and therefore an increased demand for metabolites. Thus, the right balance of each dietary component helps to maintain not only normal homeostasis but also the ability to quickly respond to systemic stress. In addition, some dietary components can drive chronic inflammatory changes in the absence of infection or immune stress, which in turn can impact on overall immune function. The optimal nutrition for the best immunological outcomes would therefore be a diet that supports the functions of immune cells allowing them to initiate effective responses against pathogens but also to resolve the response rapidly when necessary and to avoid any underlying chronic inflammation. In this review we discuss how these key dietary components can alter immune function, what is their impact on bone marrow metabolism and how changes in dietary intake of each of these can improve the outcomes of infections.
Collapse
Affiliation(s)
- Charlotte Hellmich
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
- Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Norwich, United Kingdom
| | - Edyta E. Wojtowicz
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
45
|
Benova A, Ferencakova M, Bardova K, Funda J, Prochazka J, Spoutil F, Cajka T, Dzubanova M, Balcaen T, Kerckhofs G, Willekens W, van Lenthe GH, Alquicer G, Pecinova A, Mracek T, Horakova O, Rossmeisl M, Kopecky J, Tencerova M. Novel thiazolidinedione analog reduces a negative impact on bone and mesenchymal stem cell properties in obese mice compared to classical thiazolidinediones. Mol Metab 2022; 65:101598. [PMID: 36103974 PMCID: PMC9508355 DOI: 10.1016/j.molmet.2022.101598] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022] Open
Abstract
Objective The use of thiazolidinediones (TZDs) as insulin sensitizers has been shown to have side effects including increased accumulation of bone marrow adipocytes (BMAds) associated with a higher fracture risk and bone loss. A novel TZD analog MSDC-0602K with low affinity to PPARγ has been developed to reduce adverse effects of TZD therapy. However, the effect of MSDC-0602K on bone phenotype and bone marrow mesenchymal stem cells (BM-MSCs) in relation to obesity has not been intensively studied yet. Methods Here, we investigated whether 8-week treatment with MSDC-0602K has a less detrimental effect on bone loss and BM-MSC properties in obese mice in comparison to first generation of TZDs, pioglitazone. Bone parameters (bone microstructure, bone marrow adiposity, bone strength) were examined by μCT and 3-point bending test. Primary BM-MSCs were isolated and measured for osteoblast and adipocyte differentiation. Cellular senescence, bioenergetic profiling, nutrient consumption and insulin signaling were also determined. Results The findings demonstrate that MSDC-0602K improved bone parameters along with increased proportion of smaller BMAds in tibia of obese mice when compared to pioglitazone. Further, primary BM-MSCs isolated from treated mice and human BM-MSCs revealed decreased adipocyte and higher osteoblast differentiation accompanied with less inflammatory and senescent phenotype induced by MSDC-0602K vs. pioglitazone. These changes were further reflected by increased glycolytic activity differently affecting glutamine and glucose cellular metabolism in MSDC-0602K-treated cells compared to pioglitazone, associated with higher osteogenesis. Conclusion Our study provides novel insights into the action of MSDC-0602K in obese mice, characterized by the absence of detrimental effects on bone quality and BM-MSC metabolism when compared to classical TZDs and thus suggesting a potential therapeutical use of MSDC-0602K in both metabolic and bone diseases. MSDC-0602K improves bone quality and increases proportion of smaller BMAds in obese mice. MSDC-0602K-treated mice show lower adipogenic differentiation with less senescent phenotype in primary BM-MSCs. MSDC-0602K induces higher glycolytic activity in BM-MSCs compared to pioglitazone. MSDC-0602-treated BM-MSCs prefer glutamine over glucose uptake in comparison to AT-MSCs. Beneficial effect of MSDC-06002K in BM-MSCs manifests by absence of MPC inhibition.
Collapse
Affiliation(s)
- Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Michaela Ferencakova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Jiri Funda
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics & Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Tim Balcaen
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium; Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium; Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium; Department of Materials Engineering, KU Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; Pole of Morphology, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | | | | | - Glenda Alquicer
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Alena Pecinova
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Mracek
- Laboratory of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 142 20, Czech Republic.
| |
Collapse
|
46
|
Harnett MM, Doonan J, Lumb FE, Crowe J, Damink RO, Buitrago G, Duncombe-Moore J, Wilkinson DI, Suckling CJ, Selman C, Harnett W. The parasitic worm product ES-62 protects the osteoimmunology axis in a mouse model of obesity-accelerated ageing. Front Immunol 2022; 13:953053. [PMID: 36105811 PMCID: PMC9465317 DOI: 10.3389/fimmu.2022.953053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite significant increases in human lifespan over the last century, adoption of high calorie diets (HCD) has driven global increases in type-2 diabetes, obesity and cardiovascular disease, disorders precluding corresponding improvements in healthspan. Reflecting that such conditions are associated with chronic systemic inflammation, evidence is emerging that infection with parasitic helminths might protect against obesity-accelerated ageing, by virtue of their evolution of survival-promoting anti-inflammatory molecules. Indeed, ES-62, an anti-inflammatory secreted product of the filarial nematode Acanthocheilonema viteae, improves the healthspan of both male and female C57BL/6J mice undergoing obesity-accelerated ageing and also extends median lifespan in male animals, by positively impacting on inflammatory, adipose metabolic and gut microbiome parameters of ageing. We therefore explored whether ES-62 affects the osteoimmunology axis that integrates environmental signals, such as diet and the gut microbiome to homeostatically regulate haematopoiesis and training of immune responses, which become dysregulated during (obesity-accelerated) ageing. Of note, we find sexual dimorphisms in the decline in bone health, and associated dysregulation of haematopoiesis and consequent peripheral immune responses, during obesity-accelerated ageing, highlighting the importance of developing sex-specific anti-ageing strategies. Related to this, ES-62 protects trabecular bone structure, maintaining bone marrow (BM) niches that counter the ageing-associated decline in haematopoietic stem cell (HSC) functionality highlighted by a bias towards myeloid lineages, in male but not female, HCD-fed mice. This is evidenced by the ability of ES-62 to suppress the adipocyte and megakaryocyte bias and correspondingly promote increases in B lymphocytes in the BM. Furthermore, the consequent prevention of ageing-associated myeloid/lymphoid skewing is associated with reduced accumulation of inflammatory CD11c+ macrophages and IL-1β in adipose tissue, disrupting the perpetuation of inflammation-driven dysregulation of haematopoiesis during obesity-accelerated ageing in male HCD-fed mice. Finally, we report the ability of small drug-like molecule analogues of ES-62 to mimic some of its key actions, particularly in strongly protecting trabecular bone structure, highlighting the translational potential of these studies.
Collapse
Affiliation(s)
- Margaret M. Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - James Doonan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Felicity E. Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Roel Olde Damink
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Geraldine Buitrago
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Josephine Duncombe-Moore
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Debbie I. Wilkinson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Colin J. Suckling
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
47
|
Zhao P, Xu A, Leung WK. Obesity, Bone Loss, and Periodontitis: The Interlink. Biomolecules 2022; 12:biom12070865. [PMID: 35883424 PMCID: PMC9313439 DOI: 10.3390/biom12070865] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/18/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity and periodontitis are both common health concerns that have given rise to considerable economic and societal burden worldwide. There are established negative relationships between bone metabolism and obesity, obesity and diabetes mellitus (DM), and DM and periodontitis, to name a few, with osteoporosis being considered a long-term complication of obesity. In the oral cavity, bone metabolic disorders primarily display as increased risks for periodontitis and alveolar bone loss. Obesity-driven alveolar bone loss and mandibular osteoporosis have been observed in animal models without inoculation of periodontopathogens. Clinical reports have also indicated a possible association between obesity and periodontitis. This review systematically summarizes the clinical periodontium changes, including alveolar bone loss in obese individuals. Relevant laboratory-based reports focusing on biological interlinks in obesity-associated bone remodeling via processes like hyperinflammation, immune dysregulation, and microbial dysbiosis, were reviewed. We also discuss the potential mechanism underlying obesity-enhanced alveolar bone loss from both the systemic and periodontal perspectives, focusing on delineating the practical considerations for managing periodontal disease in obese patients.
Collapse
Affiliation(s)
- Pengfei Zhao
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China;
| | - Aimin Xu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China;
| | - Wai Keung Leung
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China;
- Correspondence: ; Tel.: +852-2859-0417
| |
Collapse
|
48
|
CD4+ and CD8+ T-cell responses in bone marrow to fatty acids in high-fat diets. J Nutr Biochem 2022; 107:109057. [DOI: 10.1016/j.jnutbio.2022.109057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/23/2022] [Accepted: 04/06/2022] [Indexed: 12/30/2022]
|
49
|
Abstract
Purpose of review Immunological memory is an important evolutionary adaptation of the immune system. Previously restricted to the adaptive immune system, the concept of memory has recently been broadened to the innate immune system. This review summarizes recent studies that highlight the contribution of the hematopoietic stem cells (HSCs) in supporting immunological memory. Recent findings Short-lived innate immune cells can build a long-lasting memory of infection to improve their response to secondary challenges. Studies show that these unexpected properties of the innate immune system are sustained by epigenetic and metabolic changes in the HSC compartment. Summary HSCs are durably altered in response to pathogens and serve as long-term support for innate immune memory. Many questions remain regarding the mechanisms contributing to the induction and the maintenance of this immune memory in HSCs. Answering these questions will open new perspectives to understand how environmental factors shape the HSC activity over time.
Collapse
|
50
|
Thom CS, Wilken MB, Chou ST, Voight BF. Body mass index and adipose distribution have opposing genetic impacts on human blood traits. eLife 2022; 11:e75317. [PMID: 35166671 PMCID: PMC8884725 DOI: 10.7554/elife.75317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/14/2022] [Indexed: 12/02/2022] Open
Abstract
Body mass index (BMI), hyperlipidemia, and truncal adipose distribution concordantly elevate cardiovascular disease risks, but have unknown genetic effects on blood trait variation. Using Mendelian randomization, we define unexpectedly opposing roles for increased BMI and truncal adipose distribution on blood traits. Elevated genetically determined BMI and lipid levels decreased hemoglobin and hematocrit levels, consistent with clinical observations associating obesity and anemia. We found that lipid-related effects were confined to erythroid traits. In contrast, BMI affected multiple blood lineages, indicating broad effects on hematopoiesis. Increased truncal adipose distribution opposed BMI effects, increasing hemoglobin and blood cell counts across lineages. Conditional analyses indicated genes, pathways, and cell types responsible for these effects, including Leptin Receptor and other blood cell-extrinsic factors in adipocytes and endothelium that regulate hematopoietic stem and progenitor cell biology. Our findings identify novel roles for obesity on hematopoiesis, including a previously underappreciated role for genetically determined adipose distribution in determining blood cell formation and function.
Collapse
Affiliation(s)
- Christopher S Thom
- Division of Neonatology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Madison B Wilken
- Division of Neonatology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Stella T Chou
- Division of Hematology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Benjamin F Voight
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of MedicinePhiladelphiaUnited States
- Department of Genetics, University of Pennsylvania - Perelman School of MedicinePhiladelphiaUnited States
- Institute for Translational Medicine, University of Pennsylvania - Perelman School of MedicinePhiladelphiaUnited States
| |
Collapse
|