1
|
Torres L, Jonsson CA, Eliasson B, Forsblad-d'Elia H, Landgren AJ, Bilberg A, Gjertsson I, Larsson I, Klingberg E. A six-month weight loss intervention is associated with significant changes in serum biomarkers related to inflammation, bone and cartilage metabolism in obese patients with psoriatic arthritis and matched controls. BMC Rheumatol 2025; 9:58. [PMID: 40410839 PMCID: PMC12100911 DOI: 10.1186/s41927-025-00511-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Obesity is highly overrepresented in patients with psoriatic arthritis (PsA) and associated with increased disease activity and inferior treatment outcome. We have previously reported in 41 patients with PsA and body mass index (BMI) ≥ 33 kg/m2 that weight loss treatment with Very Low Energy Diet (VLED) resulted in a median weight loss of 18,6% and concomitantly a significant improvement in C-reactive protein (CRP) and disease activity at six months (M6). This sub-study analyzes the effects on serum biomarkers associated with inflammation, bone and cartilage metabolism in the same PsA patients and matched controls. METHODS Patients and controls received VLED treatment (640 kcal/day) during 12-16 weeks depending on baseline (BL) BMI < 40 or ≥ 40 kg/m2, followed by an energy restricted diet. Serum was collected at BL and M6, and biomarkers were measured with Magnetic Luminex® Assays and enzyme-linked immunosorbent assay (ELISA). Nonparametric statistics and paired comparison tests were used. RESULTS In the PsA patients, the following proteins were significantly reduced at M6 as compared to BL: hepatocyte growth factor (HGF) (median (first-third quartile) 327.9 (250.3-413.6) pg/mL vs. 271.3 (206.9-331.0) pg/mL, p < 0.01), vascular endothelial growth factor (VEGF) (79.6 (55.9-113.5) pg/mL vs. 69.6 (53.1-105.3) pg/mL, p = 0.01), B-cell activating factor (BAFF) (794.4 (716.4-868.3) pg/mL vs. 674.6 (613.2-790.5) pg/mL, p = 0.01) and cartilage oligomeric matrix protein (COMP) (266.1 (209.9-366.0) ng/mL vs. 217.0 (156.0-272.0) ng/mL, p < 0.01), whereas carboxyterminal telopeptide of type-1 collagen (CTX-1) was significantly increased (268.0 (196.0-378.5) pg/mL vs. 508.0 (350.0-640.0) pg/mL, p < 0.01). Similar results were found in the control group. CONCLUSIONS Weight loss was associated with reduced levels of serum biomarkers related to inflammation and cartilage degradation, and increased biomarkers for bone resorption. The study supports the strong relationship between obesity, inflammation, bone and cartilage metabolism, identifying BMI as a possible confounder for biomarker levels. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02917434, registered on September 21, 2016, retrospectively registered.
Collapse
Affiliation(s)
- Linda Torres
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Rheumatology, Sahlgrenska University Hospital, Gröna stråket 16, Gothenburg, 413 45, Sweden.
| | - Charlotte A Jonsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Björn Eliasson
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helena Forsblad-d'Elia
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gröna stråket 16, Gothenburg, 413 45, Sweden
| | - Anton J Landgren
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Research and Development Primary Health Care, Gothenburg, Södra Bohuslän, Sweden
| | - Annelie Bilberg
- Institute of Neuroscience and Physiology, Department of Health and Rehabilitation, Physiotherapy, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Occupational and Physiotherapy, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gröna stråket 16, Gothenburg, 413 45, Sweden
| | - Ingrid Larsson
- Department of Gastroenterology and Hepatology, Sahlgrenska University Hospital, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Klingberg
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gröna stråket 16, Gothenburg, 413 45, Sweden
| |
Collapse
|
2
|
Mayakrishnan V, Kannappan P, Balakarthikeyan J, Kim CY. Rodent model intervention for prevention and optimal management of sarcopenia: A systematic review on the beneficial effects of nutrients & non-nutrients and exercise to improve skeletal muscle health. Ageing Res Rev 2024; 102:102543. [PMID: 39427886 DOI: 10.1016/j.arr.2024.102543] [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: 12/28/2023] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Sarcopenia is a common musculoskeletal disorder characterized by degenerative processes and is strongly linked to an increased susceptibility to falls, fractures, physical limitations, and mortality. Several models have been used to explore therapeutic and preventative measures as well as to gain insight into the molecular mechanisms behind sarcopenia. With novel experimental methodologies emerging to design foods or novel versions of conventional foods, understanding the impact of nutrition on the prevention and management of sarcopenia has become important. This review provides a thorough assessment of the use of rodent models of sarcopenia for understanding the aging process, focusing the effects of nutrients, plant extracts, exercise, and combined interventions on skeletal muscle health. According to empirical research, nutraceuticals and functional foods have demonstrated potential benefits in enhancing physical performance. In preclinical investigations, the administration of herbal extracts and naturally occurring bioactive compounds yielded advantageous outcomes such as augmented muscle mass and strength generation. Furthermore, herbal treatments exhibited inhibitory effects on muscle atrophy and sarcopenia. A substantial body of information establishes a connection between diet and the muscle mass, strength, and functionality of older individuals. This suggests that nutrition has a major impact in both the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Vijayakumar Mayakrishnan
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Priya Kannappan
- PSG College of Arts & Science, Civil Aerodrome, Coimbatore, Tamil Nadu 641014, India
| | | | - Choon Young Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea; Department of Food and Nutrition, Yeungnam University Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
3
|
Jin SE, Kim J, Sung JH. Recent approaches of antibody therapeutics in androgenetic alopecia. Front Pharmacol 2024; 15:1434961. [PMID: 39221145 PMCID: PMC11362041 DOI: 10.3389/fphar.2024.1434961] [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: 05/19/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Therapeutic antibodies (Abs) have been anticipated as promising alternatives to conventional treatments such as topical minoxidil and oral finasteride for androgenetic alopecia (AGA). Due to the high molecular weight of typical Abs, the half-life of subcutaneous Abs exceeds 2 weeks, allowing an administration intervals of once a month or longer. Direct injection into the areas of hair loss is also feasible, potentially enhancing treatment efficacy while minimizing systemic side effects. However, therapeutic Abs are rarely developed for AGA therapy due to the requirement to be responsiveness to androgens and to exist in the extracellular fluid or cell surface surrounding the hair follicle. In this review, we introduce recent progress of antibody therapeutics in AGA targeting the prolactin receptor, Interleukin-6 receptor, C-X-C motif chemokine ligand 12, and dickkopf 1. As therapeutic Abs for AGA are still in the early stages, targets need further validation and optimization for clinical application.
Collapse
Affiliation(s)
- Su-Eon Jin
- Epi Biotech Co., Ltd., Incheon, Republic of Korea
| | - Jino Kim
- New Hair Plastic Surgery Clinic, Seoul, Republic of Korea
| | | |
Collapse
|
4
|
Yu M, Qin K, Fan J, Zhao G, Zhao P, Zeng W, Chen C, Wang A, Wang Y, Zhong J, Zhu Y, Wagstaff W, Haydon RC, Luu HH, Ho S, Lee MJ, Strelzow J, Reid RR, He TC. The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities. Genes Dis 2024; 11:101026. [PMID: 38292186 PMCID: PMC10825312 DOI: 10.1016/j.gendis.2023.04.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/18/2023] [Accepted: 04/12/2023] [Indexed: 02/01/2024] Open
Abstract
The evolutionarily conserved Wnt signaling pathway plays a central role in development and adult tissue homeostasis across species. Wnt proteins are secreted, lipid-modified signaling molecules that activate the canonical (β-catenin dependent) and non-canonical (β-catenin independent) Wnt signaling pathways. Cellular behaviors such as proliferation, differentiation, maturation, and proper body-axis specification are carried out by the canonical pathway, which is the best characterized of the known Wnt signaling paths. Wnt signaling has emerged as an important factor in stem cell biology and is known to affect the self-renewal of stem cells in various tissues. This includes but is not limited to embryonic, hematopoietic, mesenchymal, gut, neural, and epidermal stem cells. Wnt signaling has also been implicated in tumor cells that exhibit stem cell-like properties. Wnt signaling is crucial for bone formation and presents a potential target for the development of therapeutics for bone disorders. Not surprisingly, aberrant Wnt signaling is also associated with a wide variety of diseases, including cancer. Mutations of Wnt pathway members in cancer can lead to unchecked cell proliferation, epithelial-mesenchymal transition, and metastasis. Altogether, advances in the understanding of dysregulated Wnt signaling in disease have paved the way for the development of novel therapeutics that target components of the Wnt pathway. Beginning with a brief overview of the mechanisms of canonical and non-canonical Wnt, this review aims to summarize the current knowledge of Wnt signaling in stem cells, aberrations to the Wnt pathway associated with diseases, and novel therapeutics targeting the Wnt pathway in preclinical and clinical studies.
Collapse
Affiliation(s)
- Michael Yu
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kevin Qin
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Zeng
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Neurology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523475, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Annie Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Clinical Laboratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Jiamin Zhong
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
5
|
Diaz-Castro J, Toledano JM, Sanchez-Romero J, Aguilar AC, Martín-Alvarez E, Puche-Juarez M, Moreno-Fernandez J, Pinar-Gonzalez M, Prados S, Carrillo MP, Ruiz-Duran S, De Paco Matallana C, Ochoa JJ. COVID-19 and Pregnancy: A Dangerous Mix for Bone Turnover and Metabolism Biomarkers in Placenta and Colostrum. J Clin Med 2024; 13:2124. [PMID: 38610889 PMCID: PMC11012405 DOI: 10.3390/jcm13072124] [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: 02/20/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Background: In pregnant women, COVID-19 can alter the metabolic environment, cell metabolism, and oxygen supply of trophoblastic cells and, therefore, have a negative influence on essential mechanisms of fetal development. The purpose of this study was to investigate, for the first time, the effects of COVID-19 infection during pregnancy with regard to the bone turnover and endocrine function of several metabolic biomarkers in colostrum and placenta. Methods: One hundred and twenty-four pregnant mothers were recruited from three hospitals between June 2020 and August 2021 and assigned to two groups: Control group and COVID-19 group. Metabolism biomarkers were addressed in placental tissue and colostrum. Results: Lipocalin-2 and resistin levels were higher in the placenta, revealing an underlying pro-inflammatory status in the gestation period for mothers suffering from COVID-19; a decrease in GLP-1 and leptin was also observed in this group. As for adiponectin, resistin, and insulin, their concentrations showed an increase; a decrease in GLP-1, leptin, and PYY was also reported in the colostrum of mothers suffering from COVID-19 compared with the control group. Conclusions: As for bone turnover, placental samples from mothers with COVID-19 showed lower levels of OPG, while DKK-1 increased compared with the control group. Colostrum samples showed higher levels of OPG, SOST, and PTH in the COVID-19 group, a fact that could have noteworthy implications for energy metabolism, fetal skeletal development, and postnatal bone density and mineralization. Further research is needed to explain the pathogenic mechanism of COVID-19 that may affect pregnancy, so as to assess the short-term and long-term outcomes in infants' health.
Collapse
Affiliation(s)
- Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18012 Granada, Spain
| | - Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Javier Sanchez-Romero
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario ‘Virgen de la Arrixaca’, El Palmar, 30120 Murcia, Spain; (J.S.-R.); (C.D.P.M.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, 30120 Murcia, Spain
| | - Africa Caño Aguilar
- Department of Obstetrics and Gynaecology, San Cecilio Universitary Hospital, 18071 Granada, Spain
| | - Estefanía Martín-Alvarez
- Unit of Neonatology, Pediatric Service, Hospital Universitario Materno-Infantil Virgen de las Nieves, 18014 Granada, Spain
| | - Maria Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18012 Granada, Spain
| | - Maria Pinar-Gonzalez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Sonia Prados
- Department of Obstetrics and Gynaecology, San Cecilio Universitary Hospital, 18071 Granada, Spain
| | - María Paz Carrillo
- Department of Obstetrics & Gynaecology, Virgen de las Nieves University Hospital, 18014 Granada, Spain; (M.P.C.)
| | - Susana Ruiz-Duran
- Department of Obstetrics & Gynaecology, Virgen de las Nieves University Hospital, 18014 Granada, Spain; (M.P.C.)
| | - Catalina De Paco Matallana
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario ‘Virgen de la Arrixaca’, El Palmar, 30120 Murcia, Spain; (J.S.-R.); (C.D.P.M.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, 30120 Murcia, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, Biomedical Research Centre, Health Sciences Technological Park, Avenida del Conocimiento s/n, Armilla, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18012 Granada, Spain
| |
Collapse
|
6
|
Abou Azar F, Mugabo Y, Yuen S, Del Veliz S, Paré F, Rial SA, Lavoie G, Roux PP, Lim GE. Plakoglobin regulates adipocyte differentiation independently of the Wnt/β-catenin signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119690. [PMID: 38367915 DOI: 10.1016/j.bbamcr.2024.119690] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/19/2024]
Abstract
The scaffold protein 14-3-3ζ is an established regulator of adipogenesis and postnatal adiposity. We and others have demonstrated the 14-3-3ζ interactome to be diverse and dynamic, and it can be examined to identify novel regulators of physiological processes, including adipogenesis. In the present study, we sought to determine if factors that influence adipogenesis during the development of obesity could be identified in the 14-3-3ζ interactome found in white adipose tissue of lean or obese TAP-tagged-14-3-3ζ overexpressing mice. Using mass spectrometry, differences in the abundance of novel, as well as established, adipogenic factors within the 14-3-3ζ interactome could be detected in adipose tissues. One novel candidate was revealed to be plakoglobin, the homolog of the known adipogenic inhibitor, β-catenin, and herein, we report that plakoglobin is involved in adipocyte differentiation. Plakoglobin is expressed in murine 3T3-L1 cells and is primarily localized to the nucleus, where its abundance decreases during adipogenesis. Depletion of plakoglobin by siRNA inhibited adipogenesis and reduced PPARγ2 expression, and similarly, plakoglobin depletion in human adipose-derived stem cells also impaired adipogenesis and reduced lipid accumulation post-differentiation. Transcriptional assays indicated that plakoglobin does not participate in Wnt/β-catenin signaling, as its depletion did not affect Wnt3a-mediated transcriptional activity. Taken together, our results establish plakoglobin as a novel regulator of adipogenesis in vitro and highlights the ability of using the 14-3-3ζ interactome to identify potential pro-obesogenic factors.
Collapse
Affiliation(s)
- F Abou Azar
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Y Mugabo
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - S Yuen
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - S Del Veliz
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - F Paré
- Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - S A Rial
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - G Lavoie
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Québec, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - P P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Québec, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - G E Lim
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.
| |
Collapse
|
7
|
Choi N, Hwang J, Kim DY, Kim J, Song SY, Sung J. Involvement of DKK1 secreted from adipose-derived stem cells in alopecia areata. Cell Prolif 2024; 57:e13562. [PMID: 37991164 PMCID: PMC10905327 DOI: 10.1111/cpr.13562] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/11/2023] [Accepted: 10/10/2023] [Indexed: 11/23/2023] Open
Abstract
Adipose-derived stem cells (ASCs) have shown efficacy in promoting hair growth, while DKK1 inhibits the WNT pathway, which is associated with hair loss. Our study focused on investigating the expression of DKK1 in alopecia areata (AA), a condition characterised by significant increases in the DKK1 levels in human and mouse ASCs. Treatment of interferon-γ increased the expression of DKK1 via STAT3 phosphorylation in ASCs. Treatment with recombinant DKK1 resulted in a decrease of cell growth in outer root sheath cells, whereas the use of a DKK1 neutralising antibody promoted hair growth. These results indicate that ASCs secrete DKK1, playing a crucial role in the progression and development of AA. Consequently, we generated DKK1 knockout (KO) ASCs using the Crispr/Cas9 system and evaluated their hair growth-promoting effects in an AA model. The DKK1 KO in ASCs led to enhanced cell motility and reduced cellular senescence by activating the WNT signalling pathway, while it reduced the expression of inflammatory cytokines by inactivating the NF-kB pathway. As expected, the intravenous injection of DKK1-KO-ASCs in AA mice, and the treatment with a conditioned medium derived from DKK1-KO-ASCs in hair organ culture proved to be more effective compared with the use of naïve ASCs and their conditioned medium. Overall, these findings suggest that DKK1 represents a novel therapeutic target for treating AA, and cell therapy using DKK1-KO-ASCs demonstrates greater efficiency.
Collapse
Affiliation(s)
| | | | - Doo Yeong Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical SciencesYonsei UniversityIncheonSouth Korea
| | - Jino Kim
- New Hair Plastic Surgery ClinicSeoulSouth Korea
| | - Seung Yong Song
- Institute for Human Tissue Restoration, Department of Plastic and Reconstructive SurgeryYonsei University College of MedicineSeoulSouth Korea
| | - Jong‐Hyuk Sung
- Epi Biotech Co., Ltd.IncheonSouth Korea
- College of Pharmacy, Yonsei Institute of Pharmaceutical SciencesYonsei UniversityIncheonSouth Korea
| |
Collapse
|
8
|
Daamouch S, Thiele S, Hofbauer L, Rauner M. Effects of adipocyte-specific Dkk1 deletion on bone homeostasis and obesity-induced bone loss in male mice. Endocr Connect 2023; 12:e230251. [PMID: 37615386 PMCID: PMC10563648 DOI: 10.1530/ec-23-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/24/2023] [Indexed: 08/25/2023]
Abstract
The link between obesity and low bone strength has become a significant medical concern. The canonical Wnt signaling pathway is a key regulator of mesenchymal stem cell differentiation into either osteoblasts or adipocytes with active Wnt signaling promoting osteoblastogenesis. Our previous research indicated that Dickkopf-1 (Dkk1), a Wnt inhibitor, is upregulated in bone tissue in obesity and that osteoblast-derived Dkk1 drives obesity-induced bone loss. However, Dkk1 is also produced by adipocytes, but the impact of adipogenic Dkk1 on bone remodeling and its role in obesity-induced bone loss remain unclear. Thus, in this study, we investigated the influence of adipogenic Dkk1 on bone homeostasis and obesity-induced bone loss in mice. To that end, deletion of Dkk1 in adipocytes was induced by tamoxifen administration into 8-week-old male Dkk1fl/fl;AdipoQcreERT2 mice. Bone and fat mass were analyzed at 12 and 20 weeks of age. Obesity was induced in 8-week-old male Dkk1fl/fl;AdipoQcre mice with a high-fat diet (HFD) rich in saturated fats for 12 weeks. We observed that 12-week-old male mice without adipogenic Dkk1 had a significant increase in trabecular bone volume in the vertebrae and femoral bones. While histological and serological bone formation markers were not different, the number of osteoclasts and adipocytes was decreased in the vertebral bones of Dkk1fl/fl;AdipoQcre-positive mice. Despite the increased bone mass in 12-week-old male mice, at 20 weeks of age, there was no difference in the bone volume between the controls and Dkk1fl/fl;AdipoQcre-positive mice. Also, Dkk1fl/fl;AdipoQcre-positive mice were not protected from HFD-induced bone loss. Even though mRNA expression levels of Sost, another important Wnt inhibitor, in bone from Dkk1-deficient mice fed with HFD were decreased compared to Dkk1-sufficient mice on an HFD, this did not prevent the HFD-induced suppression of bone formation. In conclusion, adipogenic Dkk1 may play a transient role in bone mass regulation during adolescence, but it does not contribute to bone homeostasis or obesity-induced bone loss later in life.
Collapse
Affiliation(s)
- Souad Daamouch
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Lorenz Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Lim PJ, Marcionelli G, Srikanthan P, Ndarugendamwo T, Pinner J, Rohrbach M, Giunta C. Perturbations in fatty acid metabolism and collagen production infer pathogenicity of a novel MBTPS2 variant in Osteogenesis imperfecta. Front Endocrinol (Lausanne) 2023; 14:1195704. [PMID: 37305034 PMCID: PMC10248412 DOI: 10.3389/fendo.2023.1195704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Osteogenesis imperfecta (OI) is a heritable and chronically debilitating skeletal dysplasia. Patients with OI typically present with reduced bone mass, tendency for recurrent fractures, short stature and bowing deformities of the long bones. Mutations causative of OI have been identified in over 20 genes involved in collagen folding, posttranslational modification and processing, and in bone mineralization and osteoblast development. In 2016, we described the first X-linked recessive form of OI caused by MBTPS2 missense variants in patients with moderate to severe phenotypes. MBTPS2 encodes site-2 protease, a Golgi transmembrane protein that activates membrane-tethered transcription factors. These transcription factors regulate genes involved in lipid metabolism, bone and cartilage development, and ER stress response. The interpretation of genetic variants in MBTPS2 is complicated by the gene's pleiotropic properties; MBTPS2 variants can also cause the dermatological conditions Ichthyosis Follicularis, Atrichia and Photophobia (IFAP), Keratosis Follicularis Spinulosa Decalvans (KFSD) and Olmsted syndrome (OS) without skeletal abnormalities typical of OI. Using control and patient-derived fibroblasts, we previously identified gene expression signatures that distinguish MBTPS2-OI from MBTPS2-IFAP/KFSD and observed stronger suppression of genes involved in fatty acid metabolism in MBTPS2-OI than in MBTPS2-IFAP/KFSD; this was coupled with alterations in the relative abundance of fatty acids in MBTPS2-OI. Furthermore, we observed a reduction in collagen deposition in the extracellular matrix by MBTPS2-OI fibroblasts. Here, we extrapolate our observations in the molecular signature unique to MBTPS2-OI to infer the pathogenicity of a novel MBTPS2 c.516A>C (p.Glu172Asp) variant of unknown significance in a male proband. The pregnancy was terminated at gestational week 21 after ultrasound scans showed bowing of femurs and tibiae and shortening of long bones particularly of the lower extremity; these were further confirmed by autopsy. By performing transcriptional analyses, gas chromatography-tandem mass spectrometry-based quantification of fatty acids and immunocytochemistry on fibroblasts derived from the umbilical cord of the proband, we observed perturbations in fatty acid metabolism and collagen production similar to what we previously described in MBTPS2-OI. These findings support pathogenicity of the MBTPS2 variant p.Glu172Asp as OI-causative and highlights the value of extrapolating molecular signatures identified in multiomics studies to characterize novel genetic variants.
Collapse
Affiliation(s)
- Pei Jin Lim
- Connective Tissue Unit, Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Giulio Marcionelli
- Connective Tissue Unit, Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Pakeerathan Srikanthan
- Department of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Timothée Ndarugendamwo
- Connective Tissue Unit, Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Jason Pinner
- Centre for Clinical Genetics, Sydney Children’s Hospital, Sydney, Australia
- UNSW Medicine and Health, University of New South Wales, Sydney, Australia
| | - Marianne Rohrbach
- Connective Tissue Unit, Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Cecilia Giunta
- Connective Tissue Unit, Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Yang Z, Huang X, Zhang J, You K, Xiong Y, Fang J, Getachew A, Cheng Z, Yu X, Wang Y, Wu F, Wang N, Feng S, Lin X, Yang F, Chen Y, Wei H, Li YX. Hepatic DKK1-driven steatosis is CD36 dependent. Life Sci Alliance 2023; 6:e202201665. [PMID: 36410795 PMCID: PMC9679335 DOI: 10.26508/lsa.202201665] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is prevalent worldwide; about 25% of NAFLD silently progress into steatohepatitis, in which some of them may develop into fibrosis, cirrhosis and liver failure. However, few drugs are available for NAFLD, partly because of an incomplete understanding of its pathogenic mechanisms. Here, using in vivo and in vitro gain- and loss-of-function approaches, we identified up-regulated DKK1 plays a pivotal role in high-fat diet-induced NAFLD and its progression. Mechanistic analysis reveals that DKK1 enhances the capacity of hepatocytes to uptake fatty acids through the ERK-PPARγ-CD36 axis. Moreover, DKK1 increased insulin resistance by activating the JNK signaling, which in turn exacerbates disorders of hepatic lipid metabolism. Our finding suggests that DKK1 may be a potential therapeutic and diagnosis candidate for NAFLD and metabolic disorder progression.
Collapse
Affiliation(s)
- Zhen Yang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinping Huang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaye Zhang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai You
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Xiong
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ji Fang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Anteneh Getachew
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ziqi Cheng
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaorui Yu
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yan Wang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Feima Wu
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ning Wang
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shufen Feng
- Department of Gastroenterology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xianhua Lin
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fan Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yan Chen
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hongcheng Wei
- Department of Gastroenterology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yin-Xiong Li
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| |
Collapse
|
11
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
12
|
Chen H, Huang H, Wang Y, Zhang Y, Liu M, Lou Y, Zhang Z, Zhu D, Li P. Decreased serum Wnt antagonist levels in patients with active acromegaly. Endocr Pract 2022; 28:515-520. [PMID: 35123069 DOI: 10.1016/j.eprac.2022.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The Wnt signalling pathway is an important modulator of bone metabolism. This study aims to clarify the changes in Wnt antagonists in active and biochemically controlled acromegalic patients. METHODS We recruited 77 patients newly diagnosed with acromegaly. Of those, 41 patients with complete follow-up data were included. Thirty healthy patients matched for age, sex, and body mass index served as controls. Wnt antagonists [sclerostin (SOST), dickkopf-related protein 1 (DKK-1), and Wnt inhibitory factor 1 (WIF-1)], bone turnover markers [osteocalcin, procollagen type 1 N-terminal propeptide (P1NP), and C-terminal telopeptide of type 1 collagen (CTX)] and the bone remodelling index were investigated at baseline and posttreatment. RESULTS Acromegalic patients had higher serum osteocalcin, P1NP, and CTX and a higher bone remodelling index than controls (P < 0.01). Serum SOST, DKK-1 and WIF-1 levels were significantly decreased in patients compared to controls (all P < 0.01). Serum SOST and WIF-1 levels were negatively correlated with growth hormone levels; SOST levels were positively correlated with WIF-1. Posttreatment, serum bone turnover markers and the bone remodelling index decreased, while SOST and WIF-1 significantly increased (P < 0.05). DKK-1 levels did not change compared to baseline (P > 0.05). In biochemically controlled patients, SOST and WIF-1 levels and bone turnover markers were restored and did not differ from those of the control participants (all P > 0.05). CONCLUSION Patients with active acromegaly exhibited significantly decreased Wnt antagonist levels. The reduction in Wnt antagonists is a compensatory mechanism to counteract increased bone fragility in active acromegaly.
Collapse
Affiliation(s)
- Huan Chen
- Department of endocrinology. Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Graduate School of Peking Union Medical College, Nanjing 210008, China
| | - Hong Huang
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yijie Wang
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yan Zhang
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Mengsi Liu
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuan Lou
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ziwei Zhang
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Dalong Zhu
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China; Department of endocrinology. Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Graduate School of Peking Union Medical College, Nanjing 210008, China
| | - Ping Li
- Department of endocrinology. Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| |
Collapse
|
13
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
14
|
Abou Azar F, Lim GE. Metabolic Contributions of Wnt Signaling: More Than Controlling Flight. Front Cell Dev Biol 2021; 9:709823. [PMID: 34568323 PMCID: PMC8458764 DOI: 10.3389/fcell.2021.709823] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
The canonical Wnt signaling pathway is ubiquitous throughout the body and influences a diverse array of physiological processes. Following the initial discovery of the Wnt signaling pathway during wing development in Drosophila melanogaster, it is now widely appreciated that active Wnt signaling in mammals is necessary for the development and growth of various tissues involved in whole-body metabolism, such as brain, liver, pancreas, muscle, and adipose. Moreover, elegant gain- and loss-of-function studies have dissected the tissue-specific roles of various downstream effector molecules in the regulation of energy homeostasis. This review attempts to highlight and summarize the contributions of the Wnt signaling pathway and its downstream effectors on whole-body metabolism and their influence on the development of metabolic diseases, such as diabetes and obesity. A better understanding of the Wnt signaling pathway in these tissues may aid in guiding the development of future therapeutics to treat metabolic diseases.
Collapse
Affiliation(s)
- Frederic Abou Azar
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
15
|
Bagchi DP, MacDougald OA. Wnt Signaling: From Mesenchymal Cell Fate to Lipogenesis and Other Mature Adipocyte Functions. Diabetes 2021; 70:1419-1430. [PMID: 34155042 PMCID: PMC8336005 DOI: 10.2337/dbi20-0015] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Wnt signaling is an ancient and evolutionarily conserved pathway with fundamental roles in the development of adipose tissues. Roles of this pathway in mesenchymal stem cell fate determination and differentiation have been extensively studied. Indeed, canonical Wnt signaling is a significant endogenous inhibitor of adipogenesis and promoter of other cell fates, including osteogenesis, chondrogenesis, and myogenesis. However, emerging genetic evidence in both humans and mice suggests central roles for Wnt signaling in body fat distribution, obesity, and metabolic dysfunction. Herein, we highlight recent studies that have begun to unravel the contributions of various Wnt pathway members to critical adipocyte functions, including carbohydrate and lipid metabolism. We further explore compelling evidence of complex and coordinated interactions between adipocytes and other cell types within adipose tissues, including stromal, immune, and endothelial cells. Given the evolutionary conservation and ubiquitous cellular distribution of this pathway, uncovering the contributions of Wnt signaling to cell metabolism has exciting implications for therapeutic intervention in widespread pathologic states, including obesity, diabetes, and cancers.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
16
|
Jung SJ, Choi YJ, Park TK, Woo SE, Kim BY, Yoon JS, Jang SY. Wnt signalling inhibits adipogenesis in orbital fibroblasts from patients with Graves' orbitopathy. Br J Ophthalmol 2021; 106:1019-1027. [PMID: 34193409 DOI: 10.1136/bjophthalmol-2020-316898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/07/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND/AIMS To investigate the role of Wnt signalling in adipogenesis using an in vitro model of Graves' orbitopathy (GO). METHODS Orbital fat was obtained from patients with GO and non-GO participants for primary orbital fibroblast (OF) culture. Expression levels of Wnt5a, Wnt10b, β-catenin, phospho-β-catenin and cyclin D1 were compared between GO and non-GO OFs. These expression levels were also determined during adipogenesis of GO and non-GO OFs. The effects of a stimulator and inhibitor of Wnt signalling on adipogenesis of GO and non-GO OFs were investigated. RESULTS Western blotting analysis showed significant reductions in β-catenin and cyclin D1 and significant enhancement of phospho-β-catenin in OFs from patients with GO, compared with OFs from non-GO participants (p<0.05). Expression of Wnt5a, Wnt10b, β-catenin and cyclin D1 in OFs was highest on day 0, and then gradually declined after induction of adipogenic differentiation. The expression levels of PPARγ, C/EBPα and C/EBPβ were reduced in Wnt stimulator-treated OFs in a dose-dependent manner. Oil red O staining confirmed that a stimulator of Wnt inhibited adipogenesis in GO OFs. CONCLUSION These results indicate that Wnt signalling inhibits adipogenesis in OFs from patients with GO and non-GO participants. Further studies are required to examine the potential of Wnt signalling as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Sang Joon Jung
- Department of Ophthalmology, Korea Army Training Center, Republic of Korea Army, Nonsan-si, Republic of Korea
| | - Yeon Jeong Choi
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Sang Earn Woo
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Bo-Yeon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Jang
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| |
Collapse
|
17
|
Maiso P, Mogollón P, Ocio EM, Garayoa M. Bone Marrow Mesenchymal Stromal Cells in Multiple Myeloma: Their Role as Active Contributors to Myeloma Progression. Cancers (Basel) 2021; 13:2542. [PMID: 34067236 PMCID: PMC8196907 DOI: 10.3390/cancers13112542] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Patricia Maiso
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| |
Collapse
|
18
|
Lim PJ, Marfurt S, Lindert U, Opitz L, Ndarugendamwo T, Srikanthan P, Poms M, Hersberger M, Langhans CD, Haas D, Rohrbach M, Giunta C. Omics Profiling of S2P Mutant Fibroblasts as a Mean to Unravel the Pathomechanism and Molecular Signatures of X-Linked MBTPS2 Osteogenesis Imperfecta. Front Genet 2021; 12:662751. [PMID: 34093655 PMCID: PMC8176293 DOI: 10.3389/fgene.2021.662751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/26/2021] [Indexed: 12/03/2022] Open
Abstract
Osteogenesis imperfecta (OI) is an inherited skeletal dysplasia characterized by low bone density, bone fragility and recurrent fractures. The characterization of its heterogeneous genetic basis has allowed the identification of novel players in bone development. In 2016, we described the first X-linked recessive form of OI caused by hemizygous MBTPS2 missense variants resulting in moderate to severe phenotypes. MBTPS2 encodes site-2 protease (S2P), which activates transcription factors involved in bone (OASIS) and cartilage development (BBF2H7), ER stress response (ATF6) and lipid metabolism (SREBP) via regulated intramembrane proteolysis. In times of ER stress or sterol deficiency, the aforementioned transcription factors are sequentially cleaved by site-1 protease (S1P) and S2P. Their N-terminal fragments shuttle to the nucleus to activate gene transcription. Intriguingly, missense mutations at other positions of MBTPS2 cause the dermatological spectrum condition Ichthyosis Follicularis, Atrichia and Photophobia (IFAP) and Keratosis Follicularis Spinulosa Decalvans (KFSD) without clinical overlap with OI despite the proximity of some of the pathogenic variants. To understand how single amino acid substitutions in S2P can lead to non-overlapping phenotypes, we aimed to compare the molecular features of MBTPS2-OI and MBTPS2-IFAP/KFSD, with the ultimate goal to unravel the pathomechanisms underlying MBTPS2-OI. RNA-sequencing-based transcriptome profiling of primary skin fibroblasts from healthy controls (n = 4), MBTPS2-OI (n = 3), and MBTPS2-IFAP/KFSD (n = 2) patients was performed to identify genes that are differentially expressed in MBTPS2-OI and MBTPS2-IFAP/KFSD individuals compared to controls. We observed that SREBP-dependent genes are more downregulated in OI than in IFAP/KFSD. This is coupled to alterations in the relative abundance of fatty acids in MBTPS2-OI fibroblasts in vitro, while no consistent alterations in the sterol profile were observed. Few OASIS-dependent genes are suppressed in MBTPS2-OI, while BBF2H7- and ATF6-dependent genes are comparable between OI and IFAP/KFSD patients and control fibroblasts. Importantly, we identified genes involved in cartilage physiology that are differentially expressed in MBTPS2-OI but not in MBTPS2-IFAP/KFSD fibroblasts. In conclusion, our data provide clues to how pathogenic MBTPS2 mutations cause skeletal deformities via altered fatty acid metabolism or cartilage development that may affect bone development, mineralization and endochondral ossification.
Collapse
Affiliation(s)
- Pei Jin Lim
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| | - Severin Marfurt
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| | - Uschi Lindert
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| | - Lennart Opitz
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Timothée Ndarugendamwo
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| | - Pakeerathan Srikanthan
- University of Zürich, Zurich, Switzerland.,Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Martin Poms
- University of Zürich, Zurich, Switzerland.,Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Martin Hersberger
- University of Zürich, Zurich, Switzerland.,Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Claus-Dieter Langhans
- Department of Pediatrics, Centre for Pediatric and Adolescent Medicine, Division of Neuropediatrics and Metabolic Medicine, University Hospital, Heidelberg, Germany
| | - Dorothea Haas
- Department of Pediatrics, Centre for Pediatric and Adolescent Medicine, Division of Neuropediatrics and Metabolic Medicine, University Hospital, Heidelberg, Germany
| | - Marianne Rohrbach
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| | - Cecilia Giunta
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, Zurich, Switzerland.,University of Zürich, Zurich, Switzerland
| |
Collapse
|
19
|
Curtis KJ, Mai C, Martin H, Oberman AG, Alderfer L, Romero-Moreno R, Walsh M, Mitros SF, Thomas SG, Dynako JA, Zimmer DI, McNamara LM, Littlepage LE, Niebur GL. The effect of marrow secretome and culture environment on the rate of metastatic breast cancer cell migration in two and three dimensions. Mol Biol Cell 2021; 32:1009-1019. [PMID: 33689396 PMCID: PMC8101488 DOI: 10.1091/mbc.e19-12-0682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/21/2021] [Accepted: 03/03/2021] [Indexed: 01/01/2023] Open
Abstract
Metastasis is responsible for over 90% of cancer-related deaths, and bone is the most common site for breast cancer metastasis. Metastatic breast cancer cells home to trabecular bone, which contains hematopoietic and stromal lineage cells in the marrow. As such, it is crucial to understand whether bone or marrow cells enhance breast cancer cell migration toward the tissue. To this end, we quantified the migration of MDA-MB-231 cells toward human bone in two- and three-dimensional (3D) environments. First, we found that the cancer cells cultured on tissue culture plastic migrated toward intact trabecular bone explants at a higher rate than toward marrow-deficient bone or devitalized bone. Leptin was more abundant in conditioned media from the cocultures with intact explants, while higher levels of IL-1β, IL-6, and TNFα were detected in cultures with both intact bone and cancer cells. We further verified that the cancer cells migrated into bone marrow using a bioreactor culture system. Finally, we studied migration toward bone in 3D gelatin. Migration speed did not depend on stiffness of this homogeneous gel, but many more dendritic-shaped cancer cells oriented and migrated toward bone in stiffer gels than softer gels, suggesting a coupling between matrix mechanics and chemotactic signals.
Collapse
Affiliation(s)
- Kimberly J. Curtis
- Bioengineering Graduate Program, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| | - Christine Mai
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN 46556
| | - Hannah Martin
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN 46556
| | - Alyssa G. Oberman
- Bioengineering Graduate Program, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| | - Laura Alderfer
- Bioengineering Graduate Program, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| | - Ricardo Romero-Moreno
- Department of Chemistry and Biochemistry, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| | - Mark Walsh
- Indiana University School of Medicine, South Bend Campus, Notre Dame, IN 46556
- Beacon Medical Group, Trauma and Surgical Services, South Bend, IN, 46601
| | - Stephen F. Mitros
- Beacon Medical Group, Trauma and Surgical Services, South Bend, IN, 46601
| | - Scott G. Thomas
- Indiana University School of Medicine, South Bend Campus, Notre Dame, IN 46556
| | - Joseph A. Dynako
- Indiana University School of Medicine, South Bend Campus, Notre Dame, IN 46556
| | - David I. Zimmer
- Indiana University School of Medicine, South Bend Campus, Notre Dame, IN 46556
| | - Laoise M. McNamara
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland H91 CF50
| | - Laurie E. Littlepage
- Department of Chemistry and Biochemistry, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| | - Glen L. Niebur
- Bioengineering Graduate Program, University of Notre Dame, IN 46556
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN 46556
- Harper Cancer Research Institute, University of Notre Dame, IN 46556
| |
Collapse
|
20
|
Muoio F, Panella S, Jossen V, Lindner M, Harder Y, Müller M, Eibl R, Tallone T. Human Adipose Stem Cells (hASCs) Grown on Biodegradable Microcarriers in Serum- and Xeno-Free Medium Preserve Their Undifferentiated Status. J Funct Biomater 2021; 12:jfb12020025. [PMID: 33923488 PMCID: PMC8167760 DOI: 10.3390/jfb12020025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Human adipose stem cells (hASCs) are promising candidates for cell-based therapies, but they need to be efficiently expanded in vitro as they cannot be harvested in sufficient quantities. Recently, dynamic bioreactor systems operated with microcarriers achieved considerable high cell densities. Thus, they are a viable alternative to static planar cultivation systems to obtain high numbers of clinical-grade hASCs. Nevertheless, the production of considerable biomass in a short time must not be achieved to the detriment of the cells' quality. To facilitate the scalable expansion of hASC, we have developed a new serum- and xeno-free medium (UrSuppe) and a biodegradable microcarrier (BR44). In this study, we investigated whether the culture of hASCs in defined serum-free conditions on microcarriers (3D) or on planar (2D) cell culture vessels may influence the expression of some marker genes linked with the immature degree or the differentiated status of the cells. Furthermore, we investigated whether the biomaterials, which form our biodegradable MCs, may affect cell behavior and differentiation. The results confirmed that the quality and the undifferentiated status of the hASCs are very well preserved when they grow on BR44 MCs in defined serum-free conditions. Indeed, the ASCs showed a gene expression profile more compatible with an undifferentiated status than the same cells grown under standard planar conditions.
Collapse
Affiliation(s)
- Francesco Muoio
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Stefano Panella
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Valentin Jossen
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | | | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery, EOC, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | | | - Regine Eibl
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | - Tiziano Tallone
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
- Correspondence: ; Tel.: +41-91-805-38-85
| |
Collapse
|
21
|
Lehmann J, Thiele S, Baschant U, Rachner TD, Niehrs C, Hofbauer LC, Rauner M. Mice lacking DKK1 in T cells exhibit high bone mass and are protected from estrogen-deficiency-induced bone loss. iScience 2021; 24:102224. [PMID: 33748710 PMCID: PMC7961106 DOI: 10.1016/j.isci.2021.102224] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/16/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
The Wnt inhibitor Dickkopf-1 (DKK1) is a negative regulator of bone formation and bone mass and is dysregulated in various bone diseases. How DKK1 contributes to postmenopausal osteoporosis, however, remains poorly understood. Here, we show that mice lacking DKK1 in T cells are protected from ovariectomy-induced bone loss. Ovariectomy activated CD4+ and CD8+ T cells and increased their production of DKK1. Co-culture of activated T cells with osteoblasts inhibited Wnt signaling in osteoblasts, leading to impaired differentiation. Importantly, DKK1 expression in T cells also controlled physiological bone remodeling. T-cell-deficient Dkk1 knock-out mice had a higher bone mass with an increased bone formation rate and decreased numbers of osteoclasts compared with controls, a phenotype that was rescued by adoptive transfer of wild-type T cells. Thus, these findings highlight that T cells control bone remodeling in health and disease via their expression of DKK1.
Collapse
Affiliation(s)
- Juliane Lehmann
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
22
|
Blay C, Haffray P, Bugeon J, D’Ambrosio J, Dechamp N, Collewet G, Enez F, Petit V, Cousin X, Corraze G, Phocas F, Dupont-Nivet M. Genetic Parameters and Genome-Wide Association Studies of Quality Traits Characterised Using Imaging Technologies in Rainbow Trout, Oncorhynchus mykiss. Front Genet 2021; 12:639223. [PMID: 33692832 PMCID: PMC7937956 DOI: 10.3389/fgene.2021.639223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
One of the top priorities of the aquaculture industry is the genetic improvement of economically important traits in fish, such as those related to processing and quality. However, the accuracy of genetic evaluations has been hindered by a lack of data on such traits from a sufficiently large population of animals. The objectives of this study were thus threefold: (i) to estimate genetic parameters of growth-, yield-, and quality-related traits in rainbow trout (Oncorhynchus mykiss) using three different phenotyping technologies [invasive and non-invasive: microwave-based, digital image analysis, and magnetic resonance imaging (MRI)], (ii) to detect quantitative trait loci (QTLs) associated with these traits, and (iii) to identify candidate genes present within these QTL regions. Our study collected data from 1,379 fish on growth, yield-related traits (body weight, condition coefficient, head yield, carcass yield, headless gutted carcass yield), and quality-related traits (total fat, percentage of fat in subcutaneous adipose tissue, percentage of fat in flesh, flesh colour); genotypic data were then obtained for all fish using the 57K SNP Axiom® Trout Genotyping array. Heritability estimates for most of the 14 traits examined were moderate to strong, varying from 0.12 to 0.67. Most traits were clearly polygenic, but our genome-wide association studies (GWASs) identified two genomic regions on chromosome 8 that explained up to 10% of the genetic variance (cumulative effects of two QTLs) for several traits (weight, condition coefficient, subcutaneous and total fat content, carcass and headless gutted carcass yields). For flesh colour traits, six QTLs explained 1-4% of the genetic variance. Within these regions, we identified several genes (htr1, gnpat, ephx1, bcmo1, and cyp2x) that have been implicated in adipogenesis or carotenoid metabolism, and thus represent good candidates for further functional validation. Finally, of the three techniques used for phenotyping, MRI demonstrated particular promise for measurements of fat content and distribution, while the digital image analysis-based approach was very useful in quantifying colour-related traits. This work provides new insights that may aid the development of commercial breeding programmes in rainbow trout, specifically with regard to the genetic improvement of yield and flesh-quality traits as well as the use of invasive and/or non-invasive technologies to predict such traits.
Collapse
Affiliation(s)
- Carole Blay
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | | | - Jonathan D’Ambrosio
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
- SYSAAF, Station LPGP-INRAE, Rennes, France
| | - Nicolas Dechamp
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | | | | | - Xavier Cousin
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
- MARBEC, University of Montpellier, CNRS, Ifremer, IRD, Palavas-les-Flots, France
| | - Geneviève Corraze
- INRAE, University of Pau & Pays Adour, E2S UPPA, UMR 1419 NuMéA, Saint-Pée-sur-Nivelle, France
| | - Florence Phocas
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | |
Collapse
|
23
|
Russell T, Watad A, Bridgewood C, Rowe H, Khan A, Rao A, Loughenbury P, Millner P, Dunsmuir R, Cuthbert R, Altaie A, Jones E, McGonagle D. IL-17A and TNF Modulate Normal Human Spinal Entheseal Bone and Soft Tissue Mesenchymal Stem Cell Osteogenesis, Adipogenesis, and Stromal Function. Cells 2021; 10:cells10020341. [PMID: 33562025 PMCID: PMC7915379 DOI: 10.3390/cells10020341] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Objective: The spondylarthritides (SpA) are intimately linked to new bone formation and IL-17A and TNF pathways. We investigated spinal soft tissue and bone mesenchymal stem cell (MSC) responses to IL-17A and TNF, including their osteogenesis, adipogenesis, and stromal supportive function and ability to support lymphocyte recruitment. Methods: Normal spinal peri-entheseal bone (PEB) and entheseal soft tissue (EST) were characterized for MSCs by immunophenotypic, osteogenic, chondrogenic, and adipogenic differentiation criteria. Functional and gene transcriptomic analysis was carried out on undifferentiated, adipo- differentiated, and osteo-differentiated MSCs. The enthesis C-C Motif Chemokine Ligand 20-C-C Motif Chemokine Receptor 6 (CCL20-CCR6) axis was investigated at transcript and protein levels to ascertain whether entheseal MSCs influence local immune cell populations. Results: Cultured MSCs from both PEB and EST displayed a tri-lineage differentiation ability. EST MSCs exhibited 4.9-fold greater adipogenesis (p < 0.001) and a 3-fold lower osteogenic capacity (p < 0.05). IL-17A induced greater osteogenesis in PEB MSCs compared to EST MSCs. IL-17A suppressed adipogenic differentiation, with a significant decrease in fatty acid-binding protein 4 (FABP4), peroxisome proliferator-activated receptor gamma (PPARγ), Cell Death Inducing DFFA Like Effector C (CIDEC), and Perilipin-1 (PLIN1). IL-17A significantly increased the CCL20 transcript (p < 0.01) and protein expression (p < 0.001) in MSCs supporting a role in type 17 lymphocyte recruitment. Conclusions: Normal spinal enthesis harbors resident MSCs with different in vitro functionalities in bone and soft tissue, especially in response to IL-17A, which enhanced osteogenesis and CCL20 production and reduced adipogenesis compared to unstimulated MSCs. This MSC-stromal-enthesis immune system may be a hitherto unappreciated mechanism of “fine tuning” tissue repair responses at the enthesis in health and could be relevant for SpA understanding.
Collapse
Affiliation(s)
- Tobias Russell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
- Zabludowicz Center for Autoimmune Diseases, Department of Medicine “B”, Sheba Medical Center, Tel-Hashomer, Ramat Gan 52621, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Ramat Aviv 69978, Israel
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Hannah Rowe
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Almas Khan
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK; (A.K.); (A.R.); (P.L.); (P.M.); (R.D.)
| | - Abhay Rao
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK; (A.K.); (A.R.); (P.L.); (P.M.); (R.D.)
| | - Peter Loughenbury
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK; (A.K.); (A.R.); (P.L.); (P.M.); (R.D.)
| | - Peter Millner
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK; (A.K.); (A.R.); (P.L.); (P.M.); (R.D.)
| | - Robert Dunsmuir
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK; (A.K.); (A.R.); (P.L.); (P.M.); (R.D.)
| | - Richard Cuthbert
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Ala Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS7 4SA, UK; (T.R.); (A.W.); (C.B.); (H.R.); (R.C.); (A.A.); (E.J.)
- Correspondence: ; Tel.: +44(0)-113-392-4747
| |
Collapse
|
24
|
Camacho-Cardenosa M, Quesada-Gómez JM, Camacho-Cardenosa A, Leal A, Dorado G, Torrecillas-Baena B, Casado-Díaz A. Effects of normobaric cyclic hypoxia exposure on mesenchymal stem-cell differentiation-pilot study on bone parameters in elderly. World J Stem Cells 2020; 12:1667-1690. [PMID: 33505607 PMCID: PMC7789125 DOI: 10.4252/wjsc.v12.i12.1667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) of bone marrow are the progenitor of osteoblasts and adipocytes. MSC tend to differentiate into adipocytes, instead of osteoblasts, with aging. This favors the loss of bone mass and development of osteoporosis. Hypoxia induces hypoxia inducible factor 1α gene encoding transcription factor, which regulates the expression of genes related to energy metabolism and angiogenesis. That allows a better adaptation to low O2 conditions. Sustained hypoxia has negative effects on bone metabolism, favoring bone resorption. Yet, surprisingly, cyclic hypoxia (CH), short times of hypoxia followed by long times in normoxia, can modulate MSC differentiation and improve bone health in aging. AIM To evaluate the CH effect on MSC differentiation, and whether it improves bone mineral density in elderly. METHODS MSC cultures were induced to differentiate into osteoblasts or adipocytes, in CH (3% O2 for 1, 2 or 4 h, 4 d a week). Extracellular-matrix mineralization and lipid-droplet formation were studied in MSC induced to differentiate into osteoblast or adipocytes, respectively. In addition, gene expression of marker genes, for osteogenesis or adipogenesis, have been quantified by quantitative real time polymerase chain reaction. The in vivo studies with elderly (> 75 years old; n = 10) were carried out in a hypoxia chamber, simulating an altitude of 2500 m above sea level, or in normoxia, for 18 wk (36 CH sessions of 16 min each). Percentages of fat mass and bone mineral density from whole body, trunk and right proximal femur (femoral, femoral neck and trochanter) were assessed, using dual-energy X-ray absorptiometry. RESULTS CH (4 h of hypoxic exposure) inhibited extracellular matrix mineralization and lipid-droplet formation in MSC induced to differentiate into osteoblasts or adipocytes, respectively. However, both parameters were not significantly affected by the other shorter hypoxia times assessed. The longest periods of hypoxia downregulated the expression of genes related to extracellular matrix formation, in MSC induced to differentiate into osteoblasts. Interestingly, osteocalcin (associated to energy metabolism) was upregulated. Vascular endothelial growth factor an expression and low-density lipoprotein receptor related protein 5/6/dickkopf Wnt signaling pathway inhibitor 1 (associated to Wnt/β-catenin pathway activation) increased in osteoblasts. Yet, they decreased in adipocytes after CH treatments, mainly with the longest hypoxia times. However, the same CH treatments increased the osteoprotegerin/receptor activator for nuclear factor kappa B ligand ratio in both cell types. An increase in total bone mineral density was observed in elderly people exposed to CH, but not in specific regions. The percentage of fat did not vary between groups. CONCLUSION CH may have positive effects on bone health in the elderly, due to its possible inhibitory effect on bone resorption, by increasing the osteoprotegerin / receptor activator for nuclear factor kappa B ligand ratio.
Collapse
Affiliation(s)
| | - José Manuel Quesada-Gómez
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | | | - Alejo Leal
- Servicio de Traumatología, Hospital de Cáceres, Cáceres 10004, Spain
| | - Gabriel Dorado
- Departamento Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba-CIBERFES, 14071 Córdoba, Spain
| | - Bárbara Torrecillas-Baena
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Antonio Casado-Díaz
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| |
Collapse
|
25
|
Kim-Hellmuth S, Aguet F, Oliva M, Muñoz-Aguirre M, Kasela S, Wucher V, Castel SE, Hamel AR, Viñuela A, Roberts AL, Mangul S, Wen X, Wang G, Barbeira AN, Garrido-Martín D, Nadel BB, Zou Y, Bonazzola R, Quan J, Brown A, Martinez-Perez A, Soria JM, Getz G, Dermitzakis ET, Small KS, Stephens M, Xi HS, Im HK, Guigó R, Segrè AV, Stranger BE, Ardlie KG, Lappalainen T. Cell type-specific genetic regulation of gene expression across human tissues. Science 2020; 369:eaaz8528. [PMID: 32913075 PMCID: PMC8051643 DOI: 10.1126/science.aaz8528] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 07/31/2020] [Indexed: 12/15/2022]
Abstract
The Genotype-Tissue Expression (GTEx) project has identified expression and splicing quantitative trait loci in cis (QTLs) for the majority of genes across a wide range of human tissues. However, the functional characterization of these QTLs has been limited by the heterogeneous cellular composition of GTEx tissue samples. We mapped interactions between computational estimates of cell type abundance and genotype to identify cell type-interaction QTLs for seven cell types and show that cell type-interaction expression QTLs (eQTLs) provide finer resolution to tissue specificity than bulk tissue cis-eQTLs. Analyses of genetic associations with 87 complex traits show a contribution from cell type-interaction QTLs and enables the discovery of hundreds of previously unidentified colocalized loci that are masked in bulk tissue.
Collapse
Affiliation(s)
- Sarah Kim-Hellmuth
- Statistical Genetics, Max Planck Institute of Psychiatry, Munich, Germany.
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - François Aguet
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Meritxell Oliva
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Manuel Muñoz-Aguirre
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Catalonia, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya (UPC), Barcelona, Catalonia, Spain
| | - Silva Kasela
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Valentin Wucher
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Catalonia, Spain
| | - Stephane E Castel
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Andrew R Hamel
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ana Viñuela
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Serghei Mangul
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Gao Wang
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Alvaro N Barbeira
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Diego Garrido-Martín
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Catalonia, Spain
| | - Brian B Nadel
- Department of Molecular, Cellular, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuxin Zou
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Rodrigo Bonazzola
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jie Quan
- Inflammation & Immunology, Pfizer, Cambridge, MA, USA
| | - Andrew Brown
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Population Health and Genomics, University of Dundee, Dundee, Scotland, UK
| | - Angel Martinez-Perez
- Unit of Genomic of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - José Manuel Soria
- Unit of Genomic of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Gad Getz
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Hualin S Xi
- Foundational Neuroscience Center, AbbVie, Cambridge, MA, USA
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Ayellet V Segrè
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Barbara E Stranger
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
- Center for Genetic Medicine, Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | - Tuuli Lappalainen
- New York Genome Center, New York, NY, USA.
- Department of Systems Biology, Columbia University, New York, NY, USA
| |
Collapse
|
26
|
Bagchi DP, Nishii A, Li Z, DelProposto JB, Corsa CA, Mori H, Hardij J, Learman BS, Lumeng CN, MacDougald OA. Wnt/β-catenin signaling regulates adipose tissue lipogenesis and adipocyte-specific loss is rigorously defended by neighboring stromal-vascular cells. Mol Metab 2020; 42:101078. [PMID: 32919095 PMCID: PMC7554252 DOI: 10.1016/j.molmet.2020.101078] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/14/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Canonical Wnt/β-catenin signaling is a well-studied endogenous regulator of mesenchymal cell fate determination, promoting osteoblastogenesis and inhibiting adipogenesis. However, emerging genetic evidence in humans links a number of Wnt pathway members to body fat distribution, obesity, and metabolic dysfunction, suggesting that this pathway also functions in adipocytes. Recent studies in mice have uncovered compelling evidence that the Wnt signaling pathway plays important roles in adipocyte metabolism, particularly under obesogenic conditions. However, complexities in Wnt signaling and differences in experimental models and approaches have thus far limited our understanding of its specific roles in this context. METHODS To investigate roles of the canonical Wnt pathway in the regulation of adipocyte metabolism, we generated adipocyte-specific β-catenin (β-cat) knockout mouse and cultured cell models. We used RNA sequencing, ChIP sequencing, and molecular approaches to assess expression of Wnt targets and lipogenic genes. We then used functional assays to evaluate effects of β-catenin deficiency on adipocyte metabolism, including lipid and carbohydrate handling. In mice maintained on normal chow and high-fat diets, we assessed the cellular and functional consequences of adipocyte-specific β-catenin deletion on adipose tissues and systemic metabolism. RESULTS We report that in adipocytes, the canonical Wnt/β-catenin pathway regulates de novo lipogenesis (DNL) and fatty acid monounsaturation. Further, β-catenin mediates effects of Wnt signaling on lipid metabolism in part by transcriptional regulation of Mlxipl and Srebf1. Intriguingly, adipocyte-specific loss of β-catenin is sensed and defended by CD45-/CD31- stromal cells to maintain tissue-wide Wnt signaling homeostasis in chow-fed mice. With long-term high-fat diet, this compensatory mechanism is overridden, revealing that β-catenin deletion promotes resistance to diet-induced obesity and adipocyte hypertrophy and subsequent protection from metabolic dysfunction. CONCLUSIONS Taken together, our studies demonstrate that Wnt signaling in adipocytes is required for lipogenic gene expression, de novo lipogenesis, and lipid desaturation. In addition, adipose tissues rigorously defend Wnt signaling homeostasis under standard nutritional conditions, such that stromal-vascular cells sense and compensate for adipocyte-specific loss. These findings underscore the critical importance of this pathway in adipocyte lipid metabolism and adipose tissue function.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Akira Nishii
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ziru Li
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Jennifer B DelProposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Callie A Corsa
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Hiroyuki Mori
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Julie Hardij
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Brian S Learman
- Department of Microbiology and Immunology, University of Buffalo, Buffalo, NY, USA.
| | - Carey N Lumeng
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Bagchi DP, Li Z, Corsa CA, Hardij J, Mori H, Learman BS, Lewis KT, Schill RL, Romanelli SM, MacDougald OA. Wntless regulates lipogenic gene expression in adipocytes and protects against diet-induced metabolic dysfunction. Mol Metab 2020; 39:100992. [PMID: 32325263 PMCID: PMC7264081 DOI: 10.1016/j.molmet.2020.100992] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Obesity is a key risk factor for many secondary chronic illnesses, including type 2 diabetes and cardiovascular disease. Canonical Wnt/β-catenin signaling is established as an important endogenous inhibitor of adipogenesis. This pathway is operative in mature adipocytes; however, its roles in this context remain unclear due to complexities of Wnt signaling and differences in experimental models. In this study, we used novel cultured cell and mouse models to investigate functional roles of Wnts secreted from adipocytes. METHODS We generated adipocyte-specific Wntless (Wls) knockout mice and cultured cell models to investigate molecular and metabolic consequences of disrupting Wnt secretion from mature adipocytes. To characterize Wls-deficient cultured adipocytes, we evaluated the expression of Wnt target and lipogenic genes and the downstream functional effects on carbohydrate and lipid metabolism. We also investigated the impact of adipocyte-specific Wls deletion on adipose tissues and global glucose metabolism in mice fed normal chow or high-fat diets. RESULTS Many aspects of the Wnt signaling apparatus are expressed and operative in mature adipocytes, including the Wnt chaperone Wntless. Deletion of Wntless in cultured adipocytes results in the inhibition of de novo lipogenesis and lipid monounsaturation, likely through repression of Srebf1 (SREBP1c) and Mlxipl (ChREBP) and impaired cleavage of immature SREBP1c into its active form. Adipocyte-specific Wls knockout mice (Wls-/-) have lipogenic gene expression in adipose tissues and isolated adipocytes similar to that of controls when fed a normal chow diet. However, closer investigation reveals that a subset of Wnts and downstream signaling targets are upregulated within stromal-vascular cells of Wls-/- mice, suggesting that adipose tissues defend loss of Wnt secretion from adipocytes. Interestingly, this compensation is lost with long-term high-fat diet challenges. Thus, after six months of a high-fat diet, Wls-/- mice are characterized by decreased adipocyte lipogenic gene expression, reduced visceral adiposity, and improved glucose homeostasis. CONCLUSIONS Taken together, these studies demonstrate that adipocyte-derived Wnts regulate de novo lipogenesis and lipid desaturation and coordinate the expression of lipogenic genes in adipose tissues. In addition, we report that Wnt signaling within adipose tissues is defended, such that a loss of Wnt secretion from adipocytes is sensed and compensated for by neighboring stromal-vascular cells. With chronic overnutrition, this compensatory mechanism is lost, revealing that Wls-/- mice are resistant to diet-induced obesity, adipocyte hypertrophy, and metabolic dysfunction.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ziru Li
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Callie A Corsa
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Julie Hardij
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Hiroyuki Mori
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Brian S Learman
- Department of Microbiology and Immunology, University of Buffalo, Buffalo, NY, USA.
| | - Kenneth T Lewis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Rebecca L Schill
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Steven M Romanelli
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Pujade Busqueta L, Crocker DE, Champagne CD, McCormley MC, Deyarmin JS, Houser DS, Khudyakov JI. A blubber gene expression index for evaluating stress in marine mammals. CONSERVATION PHYSIOLOGY 2020; 8:coaa082. [PMID: 32904591 PMCID: PMC7456562 DOI: 10.1093/conphys/coaa082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/06/2020] [Accepted: 08/19/2020] [Indexed: 06/11/2023]
Abstract
Evaluating the impacts of anthropogenic disturbance on free-ranging marine mammal populations, many of which are in decline, requires robust diagnostic markers of physiological stress and health. However, circulating levels of canonical 'stress hormones' such as glucocorticoids, which are commonly used to evaluate animal health, do not capture the complexity of species-specific responses and cannot be easily measured in large, fully aquatic marine mammals. Alternatively, expression of stress-responsive genes in hormone target tissues such as blubber, the specialized subcutaneous adipose tissue that can be manually or remotely sampled from many marine mammals, may be a more informative and sensitive indicator of recent (within 24 h) exposure to stressors. We previously identified genes that were upregulated in the inner blubber of juvenile northern elephant seals during experimental stimulation of the hypothalamic-pituitary-adrenal axis. In this study, we measured baseline expression levels of a subset of these genes in inner blubber of unmanipulated juvenile elephant seals of varying physiological states and correlated them with other stress markers (body condition index, corticosteroid and thyroid hormone levels). Expression of 10 genes, including those associated with lipid metabolism (ACSL1, HMGCS2, CDO1), redox homeostasis (GPX3), adipokine signaling (ADIPOQ), lipid droplet formation (PLIN1, CIDEA) and adipogenesis (DKK1, AZGP1, TGFBI), was described by three principal components and was associated with cortisol and thyroid hormone levels. Significantly, baseline gene expression levels were predictive of circulating hormone levels, suggesting that these markers may be potential indicators of exposure to stressors in marine mammal species that are inaccessible for blood sampling. A similar approach may be used to identify species-specific stress markers in other tissues that can be sampled by remote biopsy dart from free-ranging marine mammals, such as outer blubber and skin.
Collapse
Affiliation(s)
- Laura Pujade Busqueta
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Daniel E Crocker
- Biology Department, Sonoma State University, Rohnert Park, CA 94928, USA
| | | | - Molly C McCormley
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Jared S Deyarmin
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | | | - Jane I Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
- National Marine Mammal Foundation, San Diego, CA 92106, USA
| |
Collapse
|
29
|
Jossen V, Muoio F, Panella S, Harder Y, Tallone T, Eibl R. An Approach towards a GMP Compliant In-Vitro Expansion of Human Adipose Stem Cells for Autologous Therapies. Bioengineering (Basel) 2020; 7:bioengineering7030077. [PMID: 32698363 PMCID: PMC7552624 DOI: 10.3390/bioengineering7030077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Human Adipose Tissue Stem Cells (hASCs) are a valuable source of cells for clinical applications (e.g., treatment of acute myocardial infarction and inflammatory diseases), especially in the field of regenerative medicine. However, for autologous (patient-specific) and allogeneic (off-the-shelf) hASC-based therapies, in-vitro expansion is necessary prior to the clinical application in order to achieve the required cell numbers. Safe, reproducible and economic in-vitro expansion of hASCs for autologous therapies is more problematic because the cell material changes for each treatment. Moreover, cell material is normally isolated from non-healthy or older patients, which further complicates successful in-vitro expansion. Hence, the goal of this study was to perform cell expansion studies with hASCs isolated from two different patients/donors (i.e., different ages and health statuses) under xeno- and serum-free conditions in static, planar (2D) and dynamically mixed (3D) cultivation systems. Our primary aim was I) to compare donor variability under in-vitro conditions and II) to develop and establish an unstructured, segregated growth model as a proof-of-concept study. Maximum cell densities of between 0.49 and 0.65 × 105 hASCs/cm2 were achieved for both donors in 2D and 3D cultivation systems. Cell growth under static and dynamically mixed conditions was comparable, which demonstrated that hydrodynamic stresses (P/V = 0.63 W/m3, τnt = 4.96 × 10−3 Pa) acting at Ns1u (49 rpm for 10 g/L) did not negatively affect cell growth, even under serum-free conditions. However, donor-dependent differences in the cell size were found, which resulted in significantly different maximum cell densities for each of the two donors. In both cases, stemness was well maintained under static 2D and dynamic 3D conditions, as long as the cells were not hyperconfluent. The optimal point for cell harvesting was identified as between cell densities of 0.41 and 0.56 × 105 hASCs/cm2 (end of exponential growth phase). The growth model delivered reliable predictions for cell growth, substrate consumption and metabolite production in both types of cultivation systems. Therefore, the model can be used as a basis for future investigations in order to develop a robust MC-based hASC production process for autologous therapies.
Collapse
Affiliation(s)
- Valentin Jossen
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
- Correspondence: or ; Tel.: +41-58-934-5334
| | - Francesco Muoio
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Stefano Panella
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery, Ente Ospedaliero Cantonale (EOC), 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Tiziano Tallone
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.); (T.T.)
| | - Regine Eibl
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
| |
Collapse
|
30
|
Imprinting of Mesenchymal Stromal Cell Transcriptome Persists even after Treatment in Patients with Multiple Myeloma. Int J Mol Sci 2020; 21:ijms21113854. [PMID: 32481768 PMCID: PMC7312921 DOI: 10.3390/ijms21113854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/05/2023] Open
Abstract
Introduction. Multiple myeloma (MM) is a B-cell neoplasm characterized by clonal expansion of malignant plasma cells (MM cells) in the bone-marrow (BM) compartment. BM mesenchymal stromal cells (MSC) from newly diagnosed MM patients were shown to be involved in MM pathogenesis and chemoresistance. The patients displayed a distinct transcriptome and were functionally different from healthy donors’ (HD) MSC. Our aim was to determine whether MM–MSC also contributed to relapse. Methods. We obtained and characterized patients’ MSC samples at diagnosis, two years after intensive treatment, without relapse and at relapse. Results. Transcriptomic analysis revealed differences in gene expression between HD and MM-MSC, whatever the stage of the disease. An easier differentiation towards adipogenesis at the expense of osteoblatogeneis was observed, even in patients displaying a complete response to treatment. Although their transcriptome was similar, we found that MSC from relapsed patients had an increased immunosuppressive ability, compared to those from patients in remission. Conclusion. We demonstrated that imprinting of MSC transcriptome demonstrated at diagnosis of MM, persisted even after the apparent disappearance of MM cells induced by treatment, suggesting the maintenance of a local context favorable to relapse.
Collapse
|
31
|
Colditz J, Picke AK, Hofbauer LC, Rauner M. Contributions of Dickkopf-1 to Obesity-Induced Bone Loss and Marrow Adiposity. JBMR Plus 2020; 4:e10364. [PMID: 32537550 PMCID: PMC7285751 DOI: 10.1002/jbm4.10364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/08/2020] [Accepted: 04/05/2020] [Indexed: 12/20/2022] Open
Abstract
Low bone strength in overweight individuals is a significant medical problem. One important determinant of mesenchymal stem cell fate into osteoblasts or adipocytes is the Wnt signaling pathway. We recently showed that Dickkopf‐1 (DKK1), a potent Wnt inhibitor, is upregulated in obese mice. In this study, we investigated the role of DKK1 in the pathogenesis of obesity‐induced bone loss using global and tissue‐specific KO mice. Obesity was induced in 8‐week‐old male mice with an inducible global (Rosa26‐CreERT2) or osteoprogenitor‐ (Osx–Cre‐) specific deletion of Dkk1 with a high‐fat diet (HFD) containing 60% fat. After 12 weeks, body weight, bone volume, bone fat mass, and bone turnover were assessed. Dkk1fl/fl;Rosa26‐CreERT2 mice experienced a similar increase in body weight and white fat pads as control mice. A HFD significantly reduced trabecular bone mass and the bone formation rate in Cre‐ mice and Dkk1fl/fl;Rosa26‐CreERT2 mice. Interestingly, Dkk1fl/fl;Rosa26‐CreERT2 mice were protected from HFD‐induced cortical bone loss. Furthermore, a HFD was associated with increased bone marrow fat in the femur, which was less pronounced in Dkk1fl/fl;Rosa26‐CreERT2 mice. Mice with an osteoprogenitor‐specific Dkk1 deletion showed similar results as the global knockout, showing a protection against HFD‐induced cortical bone loss and an accumulation of bone marrow fat, but a similar decrease in trabecular bone volume. In summary, DKK1 appears to contribute distinctly to cortical, but not trabecular bone loss in obesity. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Ann-Kristin Picke
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Martina Rauner
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| |
Collapse
|
32
|
Cheng YH, Dong JC, Bian Q. Small molecules for mesenchymal stem cell fate determination. World J Stem Cells 2019; 11:1084-1103. [PMID: 31875870 PMCID: PMC6904864 DOI: 10.4252/wjsc.v11.i12.1084] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/13/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells harboring self-renewal and multilineage differentiation potential that are capable of differentiating into osteoblasts, adipocytes, or chondrocytes in vitro, and regulating the bone marrow microenvironment and adipose tissue remodeling in vivo. The process of fate determination is initiated by signaling molecules that drive MSCs into a specific lineage. Impairment of MSC fate determination leads to different bone and adipose tissue-related diseases, including aging, osteoporosis, and insulin resistance. Much progress has been made in recent years in discovering small molecules and their underlying mechanisms control the cell fate of MSCs both in vitro and in vivo. In this review, we summarize recent findings in applying small molecules to the trilineage commitment of MSCs, for instance, genistein, medicarpin, and icariin for the osteogenic cell fate commitment; isorhamnetin, risedronate, and arctigenin for pro-adipogenesis; and atractylenolides and dihydroartemisinin for chondrogenic fate determination. We highlight the underlying mechanisms, including direct regulation, epigenetic modification, and post-translational modification of signaling molecules in the AMPK, MAPK, Notch, PI3K/AKT, Hedgehog signaling pathways etc. and discuss the small molecules that are currently being studied in clinical trials. The target-based manipulation of lineage-specific commitment by small molecules offers substantial insights into bone marrow microenvironment regulation, adipose tissue homeostasis, and therapeutic strategies for MSC-related diseases.
Collapse
Affiliation(s)
- Yu-Hao Cheng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Jing-Cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qin Bian
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
33
|
Role of the Wnt signalling pathway in the development of endothelial disorders in response to hyperglycaemia. Expert Rev Mol Med 2019; 21:e7. [PMID: 31796147 DOI: 10.1017/erm.2019.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Diabetes mellitus (DM) is the most common metabolic disease. A WHO report from 2016 indicates that 422 million people worldwide suffer from DM or hyperglycaemia because of impaired glucose metabolism. Chronic hyperglycaemia leads to micro- and macrovessel damage, which may result in life-threatening complications. The Wnt pathway regulates cell proliferation and survival by modulating the expression of genes that control cell differentiation. Three linked Wnt pathways have been discovered thus far: a β-catenin-dependent pathway and two pathways independent of β-catenin - the planar cell polarity pathway and calcium-dependent pathway. The Wnt pathway regulates genes associated with inflammation, cell cycle, angiogenesis, fibrinolysis and other molecular processes. AREAS COVERED This review presents the current state of knowledge regarding the contribution of the Wnt pathway to endothelial ageing under hyperglycaemic conditions and provides new insights into the molecular basis of diabetic endothelial dysfunction. CONCLUSION The β-catenin-dependent pathway is a potential target in the prophylaxis and treatment of early-stage diabetes-related vascular complications. However, the underlying molecular mechanisms remain largely undetermined and require further investigation.
Collapse
|
34
|
van Schaijik B, Tan ST, Marsh RW, Itinteang T. Expression of (pro)renin receptor and its effect on endothelial cell proliferation in infantile hemangioma. Pediatr Res 2019; 86:202-207. [PMID: 31091531 DOI: 10.1038/s41390-019-0430-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/06/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Propranolol is the preferred treatment for problematic proliferating infantile hemangioma (IH) by targeting the renin-angiotensin system (RAS) expressed by IH endothelium. (Pro)renin receptor (PRR) is a major component of the RAS associated with the canonical wnt signaling pathway. We proposed that activation of PRR by renin causes proliferation of IH. METHODS The expression of PRR in IH tissue samples was investigated using immunohistochemical (IHC) staining and NanoString analysis. NanoString analysis was also used to confirm transcriptional expression of PRR in CD34-sorted proliferating IH-derived primary cell lines. MTT assay was utilized to determine the effect of exogenous renin on the number of viable IH cells. RT-qPCR was used to determine the effect of renin on the stem cell gene expression. RESULTS NanoString analysis and IHC staining confirmed transcriptional and translational expression of PRR, which was localized to the non-endothelial and the endothelial IH cell populations. MTT assay demonstrated an increased number of viable IH cells by administration of renin and the effect was negated by the wnt receptor blocker dickkopf-1. CONCLUSION Our results present a model for renin-induced increased proliferation of IH cells through PRR acting via the wnt signaling pathway, which may account for accumulation of cells in IH during the proliferative phase of the tumor.
Collapse
Affiliation(s)
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand. .,Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Wellington, New Zealand.
| | - Reginald W Marsh
- Gillies McIndoe Research Institute, Wellington, New Zealand.,University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
35
|
Colditz J, Thiele S, Baschant U, Garbe AI, Niehrs C, Hofbauer LC, Rauner M. Osteogenic Dkk1 Mediates Glucocorticoid-Induced but Not Arthritis-Induced Bone Loss. J Bone Miner Res 2019; 34:1314-1323. [PMID: 30779862 DOI: 10.1002/jbmr.3702] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/17/2022]
Abstract
Dickkopf-1 (Dkk1) is a negative regulator of bone formation and bone mass and is deregulated in bone loss induced by arthritis and glucocorticoid (GC) exposure. However, the role of Dkk1 in these pathological processes is still unknown. Here, we used conditional Dkk1 knock-out mice to determine the role of Dkk1 produced by osteolineage cells in the development of arthritis and GC-induced bone loss. Osteoprogenitor (Osx-Cre)- and osteocyte (Dmp1-Cre)-specific knock-out mice and their Cre-negative controls were subjected to two arthritis models, K/BxN and antigen-induced arthritis. Disease induction and progression were assessed. GC-induced bone loss was induced in 25-week-old female mice by implanting prednisolone (7.5 mg) slow-release pellets for 4 weeks. Dkk1fl/fl ;Osx-Cre mice subjected to K/BxN arthritis showed mildly reduced disease severity with reduced infiltration of neutrophils and T cells into affected joints and reduced bone erosions compared with Cre-negative controls. Osteocyte-specific Dkk1 deletion did not affect disease severity or local bone erosions. However, systemic bone loss at the spine was less severe in both mouse lines. In contrast to arthritis, both lines were protected from GC-induced bone loss. Although the Cre-negative controls lost about 26% and 31% bone volume potentially caused by decreased bone formation, Cre-positive mice did not exhibit such alterations. Dkk-1 deficiency in osteolineage cells protects against GC-induced bone loss, whereas it had only minor effects in arthritis. Therefore, Dkk1 may be a promising therapeutic target especially for bone diseases in which inhibition of bone formation represents the predominant mechanism. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Annette I Garbe
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
36
|
Kim SP, Da H, Li Z, Kushwaha P, Beil C, Mei L, Xiong WC, Wolfgang MJ, Clemens TL, Riddle RC. Lrp4 expression by adipocytes and osteoblasts differentially impacts sclerostin's endocrine effects on body composition and glucose metabolism. J Biol Chem 2019; 294:6899-6911. [PMID: 30842262 DOI: 10.1074/jbc.ra118.006769] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/23/2019] [Indexed: 01/10/2023] Open
Abstract
Sclerostin exerts profound local control over bone acquisition and also mediates endocrine communication between fat and bone. In bone, sclerostin's anti-osteoanabolic activity is enhanced by low-density lipoprotein receptor-related protein 4 (Lrp4), which facilitates its interaction with the Lrp5 and Lrp6 Wnt co-receptors. To determine whether Lrp4 similarly affects sclerostin's endocrine function, we examined body composition as well as glucose and fatty acid metabolism in mice rendered deficient of Lrp4 in the adipocyte (AdΔLrp4) or the osteoblast (ObΔLrp4). AdΔLrp4 mice exhibit a reduction in adipocyte hypertrophy and improved glucose and lipid homeostasis, marked by increased glucose and insulin tolerance and reduced serum fatty acids, and mirror the effect of sclerostin deficiency on whole-body metabolism. Indeed, epistasis studies place adipocyte-expressed Lrp4 and sclerostin in the same genetic cascade that regulates adipocyte function. Intriguingly, ObΔLrp4 mice, which exhibit dramatic increases in serum sclerostin, accumulate body fat and develop impairments in glucose tolerance and insulin sensitivity despite development of a high bone mass phenotype. These data indicate that expression of Lrp4 by both the adipocyte and osteoblast is required for normal sclerostin endocrine function and that the impact of sclerostin deficiency on adipocyte physiology is distinct from the effect on osteoblast function.
Collapse
Affiliation(s)
| | - Hao Da
- From the Departments of Orthopedic Surgery and
| | - Zhu Li
- From the Departments of Orthopedic Surgery and
| | | | - Conor Beil
- From the Departments of Orthopedic Surgery and
| | - Lin Mei
- the Department of Neuroscience, Case Western Reserve University Medical School, Cleveland, Ohio 44106, and
| | - Wen-Cheng Xiong
- the Department of Neuroscience, Case Western Reserve University Medical School, Cleveland, Ohio 44106, and
| | - Michael J Wolfgang
- Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Thomas L Clemens
- From the Departments of Orthopedic Surgery and.,the Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201
| | - Ryan C Riddle
- From the Departments of Orthopedic Surgery and .,the Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201
| |
Collapse
|
37
|
van der Spoel E, Oei N, Cachucho R, Roelfsema F, Berbée JFP, Blauw GJ, Pijl H, Appelman-Dijkstra NM, van Heemst D. The 24-hour serum profiles of bone markers in healthy older men and women. Bone 2019; 120:61-69. [PMID: 30291970 DOI: 10.1016/j.bone.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/18/2022]
Abstract
The process of bone turnover displays variations over 24 h, with C-terminal cross-linked telopeptide of type 1 collagen (CTX) and osteocalcin exhibiting a nadir in the afternoon and a peak in the night. In contrast, N-terminal propeptide of type 1 procollagen (P1NP) did not display an apparent 24-hour rhythm. Other emerging novel biomarkers of bone, sclerostin and Dickkopf-related protein 1 (DKK1), are markers of osteocyte activity with limited data available regarding their 24-hour profiles. In this study, we aimed to extend available data on 24-hour profiles of CTX, osteocalcin, and P1NP and to assess the 24-hour profiles of sclerostin and DKK1 in healthy older men and women and to compare these between men and women. We measured these five bone markers in EDTA plasma collected every 4 h during 24 h in 37 healthy older men and women (range 52-76 years). Differences between time points were determined using repeated measures ANOVA and cosinor analyses were performed to determine circadian rhythmicity. The circadian rhythm of CTX was confirmed by the cosinor model, with women showing larger amplitude compared to men. Osteocalcin showed higher levels during nighttime compared to daytime in both men and women. For P1NP levels we observed a small but significant increase in the night in men. Sclerostin and DKK1 did not show a circadian rhythm, but sclerostin levels differed between time points. Because of the large intraindividual variation, DKK1 as measured in this study cannot be considered a reliable marker for diagnostic or research purposes. In conclusion, when measuring CTX, osteocalcin, P1NP, or sclerostin either in clinical practice or in a research setting, one should consider the 24-hour profiles of these bone markers.
Collapse
Affiliation(s)
- Evie van der Spoel
- Section Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Nicole Oei
- Department of Developmental Psychology (ADAPT-lab), Institute of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Ricardo Cachucho
- Section Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Section Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Ferdinand Roelfsema
- Section Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jimmy F P Berbée
- Section Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Gerard J Blauw
- Section Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Hanno Pijl
- Section Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Natasha M Appelman-Dijkstra
- Section Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Diana van Heemst
- Section Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
38
|
Ali H, Zmuda JM, Cvejkus RK, Kershaw EE, Kuipers AL, Oczypok EA, Wheeler V, Bunker CH, Miljkovic I. Wnt Pathway Inhibitor DKK1: A Potential Novel Biomarker for Adiposity. J Endocr Soc 2019; 3:488-495. [PMID: 30746507 PMCID: PMC6364619 DOI: 10.1210/js.2018-00325] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/28/2018] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence indicates that ectopic skeletal muscle adiposity may be a risk factor for type 2 diabetes (T2D), especially in persons of African ancestry. In vitro studies suggest that a Wnt pathway inhibitor, Dickkopf-related protein 1 (DKK1), plays a role in adiposity regulation and could be a biomarker for adiposity in humans. The objective of this study was to test whether serum DKK1 levels relate to adiposity measures in a cohort from an African ancestry population at high risk for T2D. Fasting serum DKK1 was measured in a sample of 159 men of African ancestry aged ≥40 years (mean age ± SD, 63.5 ± 8.2 years; mean body mass index, 27.8 ± 4.5 kg/m2). Anthropometrics included total-body and trunk adiposity measured by dual-energy x-ray absorptiometry and lower-leg skeletal muscle density measured by CT [which reflects the intramuscular adiposity content (mg/cm3)]. Serum DKK1 was positively correlated with BMI (r = 0.20; P = 0.01), waist circumference (r = 0.15; P = 0.046), DXA total-body adiposity (r = 0.24; P = 0.003), and DXA trunk adiposity (r = 0.21; P = 0.009), independent of age and height. In addition, serum DKK1 was inversely correlated with skeletal muscle density (r = −0.25; P = 0.002), independent of age, BMI, and calf muscle area. No significant correlation was found between serum DKK1 and fasting serum glucose or insulin levels or insulin resistance estimated by homeostasis model assessment. These findings suggest that higher levels of serum DKK1 may be associated with greater overall, central, and ectopic skeletal muscle adiposity. Further studies are needed to unravel the potential role of DKK1 in the regulation of adiposity in humans.
Collapse
Affiliation(s)
- Hira Ali
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph M Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan K Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Allison L Kuipers
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elizabeth A Oczypok
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victor Wheeler
- Tobago Health Studies Office, Scarborough, Tobago, Trinidad & Tobago, West Indies
| | - Clareann H Bunker
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Mazon M, Julien J, Ung RV, Picard S, Hamoudi D, Tam R, Filiatrault J, Frenette J, Mac-Way F, Carreau M. Deletion of the Fanconi Anemia C Gene in Mice Leads to Skeletal Anomalies and Defective Bone Mineralization and Microarchitecture. J Bone Miner Res 2018; 33:2007-2020. [PMID: 29989666 DOI: 10.1002/jbmr.3546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/07/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022]
Abstract
Fanconi anemia (FA) is a rare genetic disorder associated with a progressive decline in hematopoietic stem cells leading to bone marrow failure. FA is also characterized by a variety of developmental defects including short stature and skeletal malformations. More than half of children affected with FA have radial-ray abnormalities, and many patients have early onset osteopenia/osteoporosis. Although many Fanconi anemia genes have been identified and a molecular pathway defined, the underlying mechanism leading to bone defects remains elusive. To understand the role of FA genes in skeletal development and bone microarchitecture, we evaluated bone physiology during embryogenesis and in adult FancA- and FancC-deficient mice. We found that both FancA-/- and FancC-/- embryos have abnormal skeletal development shown by skeletal malformations, growth delay, and reduced bone mineralization. FancC-/- adult mice present altered bone morphology and microarchitecture with a significant decrease in cortical bone mineral density in a sex-specific manner. Mechanical testing revealed that male but not female FancC-/- mice show reduced bone strength compared with their wild-type littermates. Ex vivo cultures showed that FancA-/- and FancC-/- bone marrow-derived mesenchymal stem cells (BM MSC) have impaired differentiation capabilities together with altered gene expression profiles. Our results suggest that defective bone physiology in FA occurs in utero and possibly results from altered BM MSC function. These results provide valuable insights into the mechanism involved in FA skeletal defects. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | | | | | | | | | - Rose Tam
- CHU de Québec Research Center, Québec, Canada
| | | | - Jérôme Frenette
- CHU de Québec Research Center, Québec, Canada.,Department of Readaptation, Faculty of Medicine, Université Laval, Québec, Canada
| | - Fabrice Mac-Way
- CHU de Québec Research Center, Québec, Canada.,Division of Nephrology, Faculty and Department of Medicine, Université Laval, Québec, Canada
| | - Madeleine Carreau
- CHU de Québec Research Center, Québec, Canada.,Department of Pediatrics, Faculty of Medicine, Université Laval, Québec, Canada
| |
Collapse
|
40
|
Appelman-Dijkstra NM, Papapoulos SE. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat Rev Endocrinol 2018; 14:605-623. [PMID: 30181608 DOI: 10.1038/s41574-018-0087-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The WNT signalling pathway is a key regulator of bone metabolism, particularly bone formation, which has helped to define the role of osteocytes - the most abundant bone cells - as orchestrators of bone remodelling. Several molecules involved in the control of the WNT signalling pathway have been identified as potential targets for the development of bone-building therapeutics for patients with osteoporosis. Several of these molecules have been investigated in animal models, but only inhibitors of sclerostin (which is produced by osteocytes) have been investigated in phase III clinical studies. Here, we review the rationale for these developments and the specificity and potential off-target actions of WNT-based therapeutics. We also describe the available preclinical and clinical studies and discuss the benefits and risks of using sclerostin inhibitors for the management of patients with osteoporosis.
Collapse
|
41
|
Colditz J, Thiele S, Baschant U, Niehrs C, Bonewald LF, Hofbauer LC, Rauner M. Postnatal Skeletal Deletion of Dickkopf-1 Increases Bone Formation and Bone Volume in Male and Female Mice, Despite Increased Sclerostin Expression. J Bone Miner Res 2018; 33:1698-1707. [PMID: 29734465 DOI: 10.1002/jbmr.3463] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/16/2018] [Accepted: 04/28/2018] [Indexed: 12/14/2022]
Abstract
The Wnt antagonist Dickkopf-1 (Dkk1) is a negative regulator of osteoblast function and bone mass. However, because of the lack of appropriate models, many aspects of its role in the regulation of postnatal bone turnover and its cellular source have remained unknown. In this study, we deleted Dkk1 postnatally and in different cell types using various Cre-drivers (Rosa26-ERT2-Cre, Osx-cre, Dmp1-Cre) and assessed to which extent cells of the osteoblastic lineage contribute to the effects of Dkk1 on bone turnover and homeostasis. Female and male mice were examined at 12 weeks of age. Mice with a global or cell type-specific deletion of Dkk1 showed a two- to threefold higher bone volume compared with their Cre-negative littermates. The mineral apposition rate and the bone formation rate were increased two- to fourfold in all three mouse lines, despite a significant increase in systemic and skeletal levels of sclerostin. Dkk1 deletion further reduced the number of osteoclasts about twofold, which was accompanied by a strong decrease in the receptor activator of nuclear factor-κB ligand/osteoprotegerin mRNA ratio in femoral bone. Despite similar increases in bone mass, the deletion of Dkk1 in osterix-expressing cells reduced circulating Dkk1 significantly (males, -79%; females, -77%), whereas they were not changed in Dkk1fl/fl ;Dmp1-Cre mice. However, both lines showed significantly reduced Dkk1 mRNA levels in bone. In summary, we show that lack of Dkk1 in cells of the osteoblastic lineage leads to high bone mass with increased bone formation, despite increased levels of sclerostin. Moreover, the majority of systemic Dkk1 appears to originate from osteoprogenitors but not from mature osteoblasts or osteocytes. Nevertheless, the amount of Dkk1 produced locally by more mature osteogenic cells is sufficient to modulate bone mass. Thus, this study highlights the importance of local Wnt signaling on postnatal bone homeostasis. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Departments of Anatomy and Cell Biology and Orthopaedic Surgery, School of Medicine, Indianapolis, IN, USA
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
42
|
Grünberg JR, Elvin J, Paul A, Hedjazifar S, Hammarstedt A, Smith U. CCN5/WISP2 and metabolic diseases. J Cell Commun Signal 2018; 12:309-318. [PMID: 29247377 PMCID: PMC5842198 DOI: 10.1007/s12079-017-0437-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022] Open
Abstract
Obesity and type 2 diabetes increase worldwide at an epidemic rate. It is expected that by the year 2030 around 500 million people will have diabetes; predominantly type 2 diabetes. The CCN family of proteins has become of interest in both metabolic and other common human diseases because of their effects on mesenchymal stem cell (MSCs) proliferation and differentiation as well as being important regulators of fibrosis. We here review current knowledge of the WNT1 inducible signaling pathway protein 2 (CCN5/WISP2). It has been shown to be an important regulator of both these processes through effects on both the canonical WNT and the TGFβ pathways. It is also under normal regulation by the adipogenic commitment factor BMP4, in contrast to conventional canonical WNT ligands, and allows MSCs to undergo normal adipose cell differentiation. CCN5/WISP2 is highly expressed in, and secreted by, MSCs and is an important regulator of MSCs growth. In a transgenic mouse model overexpressing CCN5/WISP2 in the adipose tissue, we have shown that it is secreted and circulating in the blood, the mice develop hypercellular white and brown adipose tissue, have increased lean body mass and enlarged hypercellular hearts. Obese transgenic mice had improved insulin sensitivity. Interestingly, the anti-fibrotic effect of CCN5/WISP2 is protective against heart failure by inhibition of the TGFβ pathway. Understanding how CCN5/WISP2 is regulated and signals is important and may be useful for developing new treatment strategies in obesity and metabolic diseases and it can also be a target in regenerative medicine.
Collapse
Affiliation(s)
- John R Grünberg
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Johannes Elvin
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Alexandra Paul
- Department of Biology and Biological Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| |
Collapse
|
43
|
Kim SP, Frey JL, Li Z, Kushwaha P, Zoch ML, Tomlinson RE, Da H, Aja S, Noh HL, Kim JK, Hussain MA, Thorek DLJ, Wolfgang MJ, Riddle RC. Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. Proc Natl Acad Sci U S A 2017; 114:E11238-E11247. [PMID: 29229807 PMCID: PMC5748171 DOI: 10.1073/pnas.1707876115] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Sclerostin has traditionally been thought of as a local inhibitor of bone acquisition that antagonizes the profound osteoanabolic capacity of activated Wnt/β-catenin signaling, but serum sclerostin levels in humans exhibit a correlation with impairments in several metabolic parameters. These data, together with the increased production of sclerostin in mouse models of type 2 diabetes, suggest an endocrine function. To determine whether sclerostin contributes to the coordination of whole-body metabolism, we examined body composition, glucose homeostasis, and fatty acid metabolism in Sost-/- mice as well as mice that overproduce sclerostin as a result of adeno-associated virus expression from the liver. Here, we show that in addition to dramatic increases in bone volume, Sost-/- mice exhibit a reduction in adipose tissue accumulation in association with increased insulin sensitivity. Sclerostin overproduction results in the opposite metabolic phenotype due to adipocyte hypertrophy. Additionally, Sost-/- mice and those administered a sclerostin-neutralizing antibody are resistant to obesogenic diet-induced disturbances in metabolism. This effect appears to be the result of sclerostin's effects on Wnt signaling and metabolism in white adipose tissue. Since adipocytes do not produce sclerostin, these findings suggest an unexplored endocrine function for sclerostin that facilitates communication between the skeleton and adipose tissue.
Collapse
Affiliation(s)
- Soohyun P Kim
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Julie L Frey
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Zhu Li
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Priyanka Kushwaha
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Meredith L Zoch
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ryan E Tomlinson
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Hao Da
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Susan Aja
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655
- Department of Medicine, Division of Endocrinology and Metabolism, University of Massachusetts Medical School, Worcester, MA 01655
- Department of Medicine, Division of Diabetes, University of Massachusetts Medical School, Worcester, MA 01655
| | - Mehboob A Hussain
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Daniel L J Thorek
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiologic Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Cancer Molecular and Functional Imaging Program, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Michael J Wolfgang
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- Baltimore Veterans Administration Medical Center, Baltimore, MD 21201
| |
Collapse
|
44
|
Qiao GY, Dong BW, Zhu CJ, Yan CY, Chen BL. Deregulation of WNT2/FZD3/β-catenin pathway compromises the estrogen synthesis in cumulus cells from patients with polycystic ovary syndrome. Biochem Biophys Res Commun 2017; 493:847-854. [DOI: 10.1016/j.bbrc.2017.07.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 11/17/2022]
|
45
|
Matsushita K, Dzau VJ. Mesenchymal stem cells in obesity: insights for translational applications. J Transl Med 2017; 97:1158-1166. [PMID: 28414326 DOI: 10.1038/labinvest.2017.42] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022] Open
Abstract
Obesity is now a major public health problem worldwide. Lifestyle modification to reduce the characteristic excess body adiposity is important in the treatment of obesity, but effective therapeutic intervention is still needed to control what has become an obesity epidemic. Unfortunately, many anti-obesity drugs have been withdrawn from market due to adverse side effects. Bariatric surgery therefore remains the most effective therapy for severe cases, although such surgery is invasive and researchers continue to seek new control strategies for obesity. Mesenchymal stem cells (MSCs) are a major source of adipocyte generation, and studies have been conducted into the potential roles of MSCs in treating obesity. However, despite significant progress in stem cell research and its potential applications for obesity, adipogenesis is a highly complex process and the molecular mechanisms governing MSC adipogenesis remain ill defined. In particular, successful clinical application of MSCs will require extensive identification and characterization of the transcriptional regulators controlling MSC adipogenesis. Since obesity is associated with the incidence of multiple important comorbidities, an in-depth understanding of the relationship between MSC adipogenesis and the comorbidities of obesity is also necessary to evaluate the potential of effective and safe MSC-based therapies for obesity. In addition, brown adipogenesis is an attractive topic from the viewpoint of therapeutic innovation and future research into MSC-based brown adipogenesis could lead to a novel breakthrough. Ongoing stem cell studies and emerging research fields such as epigenetics are expected to elucidate the complicated mechanisms at play in MSC adipogenesis and develop novel MSC-based therapeutic options for obesity. This review discusses the current understanding of MSCs in adipogenesis and their potential clinical applications for obesity.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Division of Cardiology, Second Department of Internal Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
46
|
Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia 2017; 32:575-587. [PMID: 28816238 PMCID: PMC5843902 DOI: 10.1038/leu.2017.259] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022]
Abstract
Little is known about how leukemia cells alter the bone marrow (BM) niche to facilitate their own growth and evade chemotherapy. Here, we provide evidence that acute myeloid leukemia (AML) blasts remodel the BM niche into a leukemia growth-permissive and normal hematopoiesis-suppressive microenvironment through exosome secretion. Either engrafted AML cells or AML-derived exosomes increased mesenchymal stromal progenitors and blocked osteolineage development and bone formation in vivo. Preconditioning with AML-derived exosomes ‘primed’ the animals for accelerated AML growth. Conversely, disruption of exosome secretion in AML cells through targeting Rab27a, an important regulator involved in exosome release, significantly delayed leukemia development. In BM stromal cells, AML-derived exosomes induced the expression of DKK1, a suppressor of normal hematopoiesis and osteogenesis, thereby contributing to osteoblast loss. Conversely, treatment with a DKK1 inhibitor delayed AML progression and prolonged survival in AML-engrafted mice. In addition, AML-derived exosomes induced a broad downregulation of hematopoietic stem cell-supporting factors (for example, CXCL12, KITL and IGF1) in BM stromal cells and reduced their ability to support normal hematopoiesis. Altogether, this study uncovers novel features of AML pathogenesis and unveils how AML cells create a self-strengthening leukemic niche that promotes leukemic cell proliferation and survival, while suppressing normal hematopoiesis through exosome secretion.
Collapse
|
47
|
Fairfield H, Rosen CJ, Reagan MR. Connecting Bone and Fat: The Potential Role for Sclerostin. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:114-121. [PMID: 28580233 PMCID: PMC5448707 DOI: 10.1007/s40610-017-0057-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sclerostin (SOST), a protein secreted from mature osteocytes in response to mechanical unloading and other stimuli, inhibits the osteogenic Wnt/β-catenin pathway in mesenchymal stem cells (MSCs) impeding their ability to differentiate into mineralizing osteoblasts. PURPOSE This review summarizes the crosstalk between adipose tissue and bone. It also reviews the origin, regulation, and role of SOST in osteogenesis and brings attention to an emerging role of this protein in the regulation of adipogenesis. RECENT FINDINGS Bone-derived molecules that drive MSC adipogenesis have not previously been identified, but recent findings suggest that SOST signaling may induce adipogenesis. In vivo SOST acts locally to induce changes in bone and, in vitro, increases adipogenesis in 3T3-L1 preadipocytes. SUMMARY SOST is able to induce adipogenesis in certain preadipocytes, however bone-specific studies are needed to determine the effect of local SOST concentrations in healthy and disease models on bone marrow adipose tissue.
Collapse
Affiliation(s)
- Heather Fairfield
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
| | - Clifford J. Rosen
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| | - Michaela R. Reagan
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
48
|
Mota de Sá P, Richard AJ, Hang H, Stephens JM. Transcriptional Regulation of Adipogenesis. Compr Physiol 2017; 7:635-674. [PMID: 28333384 DOI: 10.1002/cphy.c160022] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adipocytes are the defining cell type of adipose tissue. Once considered a passive participant in energy storage, adipose tissue is now recognized as a dynamic organ that contributes to several important physiological processes, such as lipid metabolism, systemic energy homeostasis, and whole-body insulin sensitivity. Therefore, understanding the mechanisms involved in its development and function is of great importance. Adipocyte differentiation is a highly orchestrated process which can vary between different fat depots as well as between the sexes. While hormones, miRNAs, cytoskeletal proteins, and many other effectors can modulate adipocyte development, the best understood regulators of adipogenesis are the transcription factors that inhibit or promote this process. Ectopic expression and knockdown approaches in cultured cells have been widely used to understand the contribution of transcription factors to adipocyte development, providing a basis for more sophisticated in vivo strategies to examine adipogenesis. To date, over two dozen transcription factors have been shown to play important roles in adipocyte development. These transcription factors belong to several families with many different DNA-binding domains. While peroxisome proliferator-activated receptor gamma (PPARγ) is undoubtedly the most important transcriptional modulator of adipocyte development in all types of adipose tissue, members of the CCAAT/enhancer-binding protein, Krüppel-like transcription factor, signal transducer and activator of transcription, GATA, early B cell factor, and interferon-regulatory factor families also regulate adipogenesis. The importance of PPARγ activity is underscored by several covalent modifications that modulate its activity and its ability to modulate adipocyte development. This review will primarily focus on the transcriptional control of adipogenesis in white fat cells and on the mechanisms involved in this fine-tuned developmental process. © 2017 American Physiological Society. Compr Physiol 7:635-674, 2017.
Collapse
Affiliation(s)
- Paula Mota de Sá
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Allison J Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Hardy Hang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
49
|
SNP-SNP interactions between WNT4 and WNT5A were associated with obesity related traits in Han Chinese Population. Sci Rep 2017; 7:43939. [PMID: 28272483 PMCID: PMC5341019 DOI: 10.1038/srep43939] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/31/2017] [Indexed: 12/02/2022] Open
Abstract
Considering the biological roles of WNT4 and WNT5A involved in adipogenesis, we aimed to investigate whether SNPs in WNT4 and WNT5A contribute to obesity related traits in Han Chinese population. Targeted genomic sequence for WNT4 and WNT5A was determined in 100 Han Chinese subjects and tag SNPs were selected. Both single SNP and SNP × SNP interaction association analyses with body mass index (BMI) were evaluated in the 100 subjects and another independent sample of 1,627 Han Chinese subjects. Meta-analyses were performed and multiple testing corrections were carried out using the Bonferroni method. Consistent with the Genetic Investigation of ANthropometric Traits (GIANT) dataset results, we didn’t detect significant association signals in single SNP association analyses. However, the interaction between rs2072920 and rs11918967, was associated with BMI after multiple testing corrections (combined P = 2.20 × 10−4). The signal was also significant in each contributing data set. SNP rs2072920 is located in the 3′-UTR of WNT4 and SNP rs11918967 is located in the intron of WNT5A. Functional annotation results revealed that both SNPs might be involved in transcriptional regulation of gene expression. Our results suggest that a combined effect of SNPs via WNT4-WNT5A interaction may affect the variation of BMI in Han Chinese population.
Collapse
|
50
|
Fulzele K, Lai F, Dedic C, Saini V, Uda Y, Shi C, Tuck P, Aronson JL, Liu X, Spatz JM, Wein MN, Pajevic PD. Osteocyte-Secreted Wnt Signaling Inhibitor Sclerostin Contributes to Beige Adipogenesis in Peripheral Fat Depots. J Bone Miner Res 2017; 32:373-384. [PMID: 27653320 PMCID: PMC5525330 DOI: 10.1002/jbmr.3001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/17/2016] [Accepted: 09/20/2016] [Indexed: 11/11/2022]
Abstract
Cells of the osteoblast lineage are increasingly identified as participants in whole-body metabolism by primarily targeting pancreatic insulin secretion or consuming energy. Osteocytes, the most abundant bone cells, secrete a Wnt-signaling inhibitor called sclerostin. Here we examined three mouse models expressing high sclerostin levels, achieved through constitutive or inducible loss of the stimulatory subunit of G-proteins (Gsα in mature osteoblasts and/or osteocytes). These mice showed progressive loss of white adipose tissue (WAT) with tendency toward increased energy expenditure but no changes in glucose or insulin metabolism. Interestingly beige adipocytes were increased extensively in both gonadal and inguinal WAT and had reduced canonical β-catenin signaling. To determine if sclerostin directly contributes to the increased beige adipogenesis, we engineered an osteocytic cell line lacking Gsα which has high sclerostin secretion. Conditioned media from these cells significantly increased expression of UCP1 in primary adipocytes, and this effect was partially reduced after depletion of sclerostin from the conditioned media. Similarly, treatment of Gsα-deficient animals with sclerostin-neutralizing antibody partially reduced the increased UCP1 expression in WAT. Moreover, direct treatment of sclerostin to wild-type mice significantly increased UCP1 expression in WAT. These results show that osteocytes and/or osteoblasts secrete factors regulating beige adipogenesis, at least in part, through the Wnt-signaling inhibitor sclerostin. Further studies are needed to assess metabolic effects of sclerostin on adipocytes and other metabolic tissues. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Keertik Fulzele
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Forest Lai
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Christopher Dedic
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Vaibhav Saini
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuhei Uda
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Chao Shi
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Padrig Tuck
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jenna L Aronson
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaolong Liu
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jordan M Spatz
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paola Divieti Pajevic
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| |
Collapse
|