1
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
3
|
De Francesco F, Sbarbati A, Sierra LAQ, Zingaretti N, Sarmadian Z, Parodi PC, Ricci G, Riccio M, Mobasheri A. Anatomy, Histology, and Embryonic Origin of Adipose Tissue: Insights to Understand Adipose Tissue Homofunctionality in Regeneration and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:53-78. [PMID: 39107527 DOI: 10.1007/5584_2024_801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Preadipocytes are formed during the 14th and 16th weeks of gestation. White adipose tissue, in particular, is generated in specific areas and thereby assembles after birth, rapidly increasing following the propagation of adipoblasts, which are considered the preadipocyte cell precursors. The second trimester of gestation is a fundamental phase of adipogenesis, and in the third trimester, adipocytes, albeit small may be present within the main deposition areas. In the course of late gestation, adipose tissue develops in the foetus and promotes the synthesis of large amounts of uncoupling protein 1, in similar quantities relative to differentiated brown adipose tissue. In mammals, differentiation occurs in two functionally different types of adipose cells: white adipose cells resulting from lipid storage and brown adipose cells from increased metabolic energy consumption. During skeletogenesis, synovial joints develop through the condensation of mesenchymal cells, which forms an insertional layer of flattened cells that umlaut skeletal elements, by sharing the same origin in the development of synovium. Peri-articular fat pads possess structural similarity with body subcutaneous white adipose tissue; however, they exhibit a distinct metabolic function due to the micro-environmental cues in which they are embedded. Fat pads are an important component of the synovial joint and play a key role in the maintenance of joint homeostasis. They are also implicated in pathological states such as osteoarthritis.In this paper we explore the therapeutic potential of adipocyte tissue mesenchymal precursor-based stem cell therapy linking it back to the anatomic origin of adipose tissue.
Collapse
Affiliation(s)
- Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona, Italy
| | | | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Zahra Sarmadian
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Joint Surgery, Sun Yat-sen University, Guangzhou, People's Republic of China.
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| |
Collapse
|
4
|
Muffová B, Králová Lesná I, Poledne R. Physiology and Pathobiology of Perivascular Adipose Tissue: Inflammation-based Atherogenesis. Physiol Res 2024; 73:929-941. [PMID: 39903884 PMCID: PMC11835208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/20/2024] [Indexed: 02/06/2025] Open
Abstract
Perivascular adipose tissue (PVAT) envelops the majority of systemic vessels, providing crucial mechanical support and vessel protection. In physiological conditions, PVAT releases various bioactive molecules, contributing to the anti-inflammatory environment around neighboring vessels. However, in conditions like obesity, PVAT can exacerbate cardiovascular issues such as atherosclerosis. Communication between PVAT and nearby vessels is bidirectional, with PVAT responding dynamically to signals from the vasculature. This responsiveness positions PVAT as a promising indicator of vascular inflammation. Recently, the role of PVAT in the CVD risk prediction is also greatly discussed. The objective of this review is to summarize the current state of knowledge about the PVAT function, its role in physiologic and pathophysiologic processes and its potential in CVD risk prediction. Keywords: Perivascular adipose tissue, inflammation, atherogenesis, Fat attenuation index.
Collapse
Affiliation(s)
- B Muffová
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | |
Collapse
|
5
|
Thakur P, Baraskar K, Shrivastava VK, Medhi B. Cross-talk between adipose tissue and microbiota-gut-brain-axis in brain development and neurological disorder. Brain Res 2024; 1844:149176. [PMID: 39182900 DOI: 10.1016/j.brainres.2024.149176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
Collapse
Affiliation(s)
- Pratibha Thakur
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India.
| | - Kirti Baraskar
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Vinoy K Shrivastava
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab 160012, India.
| |
Collapse
|
6
|
Widgerow AD. Adipose Tissue, Regeneration, and Skin Health: The Next Regenerative Frontier. Aesthet Surg J Open Forum 2024; 6:ojae117. [PMID: 39703369 PMCID: PMC11658414 DOI: 10.1093/asjof/ojae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Adipose tissue, or fat compartments, has long been considered a storage depot and an energy source. However, a large part of new research, starting with the discovery of adipose-derived stem cells, has redirected this thinking toward the tremendous regenerative capacity that adipose tissue possesses when it is healthy. This has resulted in multiple technologies being explored with fat as a basis or with fat as a target aiming at the stimulation of new small hyperplastic adipose cells exuding adipokines and encouraging the proliferation of a whole host of progenitor cells that can have positive effects on many organ systems. One of these organ systems is skin, and there is a direct correlation with various fat compartments and skin health. Dermal fat tissue, also known as dermal white adipose tissue, is one such compartment that originates from dermal preadipocytes transdifferentiating into adipocytes and progenitor adipose cells under the right cues. The author of this paper discusses these potential cues, including injectable fillers, fat grafts, and topical formulations, and their capacity to impact skin health through the generation of healthy fat tissue. In addition, small molecules such as glucagon-like peptide-1 peptides and their impact on fat tissue are discussed. Adipose tissue is being recognized as the next regenerative frontier with exciting prospects ahead. Level of Evidence 5 Therapeutic
Collapse
Affiliation(s)
- Alan D Widgerow
- Corresponding Author: Dr Alan D. Widgerow, 9 Waterway, Irvine, CA 92614, USA. E-mail: ; Instagram: @alanwidge
| |
Collapse
|
7
|
Wojciuk B, Frulenko I, Brodkiewicz A, Kita D, Baluta M, Jędrzejczyk F, Budkowska M, Turkiewicz K, Proia P, Ciechanowicz A, Kostrzewa-Nowak D, Nowak R. The Complement System as a Part of Immunometabolic Post-Exercise Response in Adipose and Muscle Tissue. Int J Mol Sci 2024; 25:11608. [PMID: 39519159 PMCID: PMC11545998 DOI: 10.3390/ijms252111608] [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] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The precise molecular processes underlying the complement's activation, which follows exposure to physical stress still remain to be fully elucidated. However, some possible mechanisms could play a role in initiating changes in the complement's activity, which are observed post-exposure to physical stress stimuli. These are mainly based on metabolic shifts that occur in the microenvironment of muscle tissue while performing its function with increased intensity, as well as the adipose tissue's role in sterile inflammation and adipokine secretion. This review aims to discuss the current opinions on the possible link between the complement activation and diet, age, sex, and health disorders with a particular emphasis on endocrinopathies and, furthermore, the type of physical activity and overall physical fitness. It has been indicated that regular physical activity incorporated into therapeutic strategies potentially improves the management of particular diseases, such as, e.g., autoimmune conditions. Moreover, it represents a favorable influence on immunoaging processes. A better understanding of the complement system's interaction with physical activity will support established clinical therapies targeting complement components.
Collapse
Affiliation(s)
- Bartosz Wojciuk
- Department of Diagnostic Immunology, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Ignacy Frulenko
- Pomeranian Medical University in Szczecin, 1 Rybacka St., 70-204 Szczecin, Poland;
- Department of Pathology, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej St., 71-242 Szczecin, Poland
| | - Andrzej Brodkiewicz
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Dagmara Kita
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Monica Baluta
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Filip Jędrzejczyk
- Department of Pediatrics, Pediatric Nephrology, Dialysis and Acute Intoxications, Pomeranian Medical University, 4 Mączna St., 70-204 Szczecin, Poland; (A.B.); (D.K.); (M.B.); (F.J.)
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University of Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Karolina Turkiewicz
- Department of Laboratory Diagnostics, University Clinical Hospital No. 2, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Patrizia Proia
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90144 Palermo, Italy;
| | - Andrzej Ciechanowicz
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
| | - Dorota Kostrzewa-Nowak
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 72 Powstańców Wlkp. Al., 70-111 Szczecin, Poland;
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, 107 Hallera St., 80-416 Gdańsk, Poland
| | - Robert Nowak
- Department of Pathology, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej St., 71-242 Szczecin, Poland
- Institute of Physical Culture Sciences, University of Szczecin, 17C Narutowicza St., 70-240 Szczecin, Poland
| |
Collapse
|
8
|
Kawasaki M, Kawasaki K, Sari FT, Kudo T, Nihara J, Kitamura M, Nagai T, Utama V, Ishida Y, Meguro F, Kesuma A, Fujita A, Nishimura T, Kogure Y, Maruyama S, Tanuma JI, Kakihara Y, Maeda T, Ghafoor S, Khonsari RH, Corre P, Sharpe PT, Cobourne M, Franco B, Ohazama A. Cell-cell interaction determines cell fate of mesoderm-derived cell in tongue development through Hh signaling. eLife 2024; 13:e85042. [PMID: 39392396 PMCID: PMC11469673 DOI: 10.7554/elife.85042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Dysfunction of primary cilia leads to genetic disorder, ciliopathies, which shows various malformations in many vital organs such as brain. Multiple tongue deformities including cleft, hamartoma, and ankyloglossia are also seen in ciliopathies, which yield difficulties in fundamental functions such as mastication and vocalization. Here, we found these tongue anomalies in mice with mutation of ciliary protein. Abnormal cranial neural crest-derived cells (CNCC) failed to evoke Hh signal for differentiation of mesoderm-derived cells into myoblasts, which resulted in abnormal differentiation of mesoderm-derived cells into adipocytes. The ectopic adipose subsequently arrested tongue swelling formation. Ankyloglossia was caused by aberrant cell migration due to lack of non-canonical Wnt signaling. In addition to ciliopathies, these tongue anomalies are often observed as non-familial condition in human. We found that these tongue deformities could be reproduced in wild-type mice by simple mechanical manipulations to disturb cellular processes which were disrupted in mutant mice. Our results provide hints for possible future treatment in ciliopathies.
Collapse
Affiliation(s)
- Maiko Kawasaki
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Katsushige Kawasaki
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Finsa Tisna Sari
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takehisa Kudo
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Jun Nihara
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Madoka Kitamura
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takahiro Nagai
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Vanessa Utama
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yoko Ishida
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Fumiya Meguro
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Alex Kesuma
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Akira Fujita
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takayuki Nishimura
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yuan Kogure
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Satoshi Maruyama
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Jun-ichi Tanuma
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yoshito Kakihara
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Sarah Ghafoor
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Roman H Khonsari
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Pierre Corre
- Service de Chirurgie Maxillofaciale et tomatology, Centre Hospitalier Universitaire de Nantes,1 place Alexis Ricordeau 44000NantesFrance
| | - Paul T Sharpe
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Martyn Cobourne
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine (TIGEM), PozzuoliNaplesItaly
- Medical Genetics, Department of Translational Medical Sciences, Federico II University of Naples, ItalyNaplesItaly
- Scuola Superiore Meridionale, School for Advanced Studies, Genomics and Experimental Medicine program,NaplesItaly
| | - Atsushi Ohazama
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| |
Collapse
|
9
|
Gruel N, Quignot C, Lesage L, El Zein S, Bonvalot S, Tzanis D, Ait Rais K, Quinquis F, Manciot B, Vibert J, El Tannir N, Dahmani A, Derrien H, Decaudin D, Bièche I, Courtois L, Mariani O, Linares LK, Gayte L, Baulande S, Waterfall JJ, Delattre O, Pierron G, Watson S. Cellular origin and clonal evolution of human dedifferentiated liposarcoma. Nat Commun 2024; 15:7941. [PMID: 39266532 PMCID: PMC11393420 DOI: 10.1038/s41467-024-52067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
Dedifferentiated liposarcoma (DDLPS) is the most frequent high-grade soft tissue sarcoma subtype. It is characterized by a component of undifferentiated tumor cells coexisting with a component of well-differentiated adipocytic tumor cells. Both dedifferentiated (DD) and well-differentiated (WD) components exhibit MDM2 amplification, however their cellular origin remains elusive. Using single-cell RNA sequencing, DNA sequencing, in situ multiplex immunofluorescence and functional assays in paired WD and DD components from primary DDLPS tumors, we characterize the cellular heterogeneity of DDLPS tumor and micro-environment. We identify a population of tumor adipocyte stem cells (ASC) showing striking similarities with adipocyte stromal progenitors found in white adipose tissue. We show that tumor ASC harbor the ancestral genomic alterations of WD and DD components, suggesting that both derive from these progenitors following clonal evolution. Last, we show that DD tumor cells keep important biological properties of ASC including pluripotency and that their adipogenic properties are inhibited by a TGF-β-high immunosuppressive tumor micro-environment.
Collapse
Affiliation(s)
- Nadège Gruel
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
- Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Chloé Quignot
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
| | - Laëtitia Lesage
- Department of Pathology, Institut Curie Hospital, Paris, France
| | - Sophie El Zein
- Department of Pathology, Institut Curie Hospital, Paris, France
| | - Sylvie Bonvalot
- Department of Surgical Oncology, Institut Curie Hospital, Paris, France
| | - Dimitri Tzanis
- Department of Surgical Oncology, Institut Curie Hospital, Paris, France
| | | | - Fabien Quinquis
- Department of Genetics, Institut Curie Hospital, Paris, France
| | - Bastien Manciot
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
| | - Julien Vibert
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
- Drug Development Department, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Nadine El Tannir
- Medico Scientific Program for Adult sarcomas, Institut Curie Research Center, Paris, France
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Department of translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Héloïse Derrien
- Laboratory of Preclinical Investigation, Department of translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Department of translational Research, PSL Research University, Institut Curie Research Center, Paris, France
- Department of Medical Oncology, Institut Curie Hospital, Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie Hospital, Paris, France
| | - Laura Courtois
- Department of Genetics, Institut Curie Hospital, Paris, France
| | - Odette Mariani
- Department of Pathology, Institut Curie Hospital, Paris, France
| | - Laëtitia K Linares
- INSERM U1194, Metabolism and Sarcoma, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, Montpellier, France
| | - Laurie Gayte
- INSERM U1194, Metabolism and Sarcoma, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, Montpellier, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, PSL Research University, Institut Curie, Paris, France
| | - Joshua J Waterfall
- Department of Translational Research, Institut Curie Research Center, Paris, France
- INSERM U830, Integrative Functional Genomics of Cancer Lab, PSL Research University, Institut Curie Research Center, Paris, France
| | - Olivier Delattre
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
- Department of Genetics, Institut Curie Hospital, Paris, France
- SIREDO Pediatric Oncology Center, Institut Curie Hospital, Paris, France
| | - Gaëlle Pierron
- Department of Genetics, Institut Curie Hospital, Paris, France
| | - Sarah Watson
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France.
- Department of Medical Oncology, Institut Curie Hospital, Paris, France.
| |
Collapse
|
10
|
Branquinho J, Neves RL, Martin RP, Arata JG, Bittencourt CA, Araújo RC, Icimoto MY, Pesquero JB. Kinin B1 receptor deficiency promotes enhanced adipose tissue thermogenic response to β3-adrenergic stimulation. Inflamm Res 2024; 73:1565-1579. [PMID: 39017739 DOI: 10.1007/s00011-024-01917-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE AND DESIGN Kinin B1 receptor (B1R) has a key role in adipocytes to protect against obesity and glycemic metabolism, thus becoming a potential target for regulation of energy metabolism and adipose tissue thermogenesis. MATERIAL OR SUBJECTS Kinin B1 knockout mice (B1KO) were subjected to acute induction with CL 316,243 and chronic cold exposure. METHODS Metabolic and histological analyses, gene and protein expression and RNA-seq were performed on interscapular brown adipose tissue (iBAT) and inguinal white adipose tissue (iWAT) of mice. RESULTS B1KO mice, under acute effect of CL 316,243, exhibited increased energy expenditure and upregulated thermogenic genes in iWAT. They were also protected from chronic cold, showing enhanced non-shivering thermogenesis with increased iBAT mass (~ 90%) and recruitment of beige adipocytes in iWAT (~ 50%). Positive modulation of thermogenic and electron transport chain genes, reaching a 14.5-fold increase for Ucp1 in iWAT. RNA-seq revealed activation of the insulin signaling pathways for iBAT and oxidative phosphorylation, tricarboxylic acid cycle, and browning pathways for iWAT. CONCLUSION B1R deficiency induced metabolic and gene expression alterations in adipose tissue, activating thermogenic pathways and increasing energy metabolism. B1R antagonists emerge as promising therapeutic targets for regulating obesity and associated metabolic disorders, such as inflammation and diabetes.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Cold Temperature
- Dioxoles/pharmacology
- Energy Metabolism/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Thermogenesis/drug effects
- Thiazoles/pharmacology
- Uncoupling Protein 1/genetics
- Uncoupling Protein 1/metabolism
Collapse
Affiliation(s)
- Jéssica Branquinho
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Raquel L Neves
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Renan P Martin
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Júlia G Arata
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Clarissa A Bittencourt
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ronaldo C Araújo
- Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Marcelo Y Icimoto
- Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil.
| | - João B Pesquero
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Gavin KM, Conrad B, Sullivan T, Vianzon R, Acosta AS, Kohrt WM, Klemm DJ. Adipogenic differentiation of hematopoietic lineage cells isolated from adipose tissue of humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.22.600230. [PMID: 38979365 PMCID: PMC11230218 DOI: 10.1101/2024.06.22.600230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
We previously discovered some adipocytes in the major white fat depots of mice and humans arise from bone marrow-derived cells of hematopoietic lineage rather than conventional mesenchymal precursors, termed bone marrow-derived adipocytes (BMDA). Here we aimed to determine if hematopoietic lineage cells isolated from adipose tissue and circulation of humans could undergo adipogenic differentiation in vitro, thereby establishing an in vitro model for studies of BMDA. We hypothesized that hematopoietic lineage cells isolated from adipose tissue, but not circulation, of humans would demonstrate adipogenic potential. Participants included younger (20-50 years) and older (>50-75 years) men and women, BMI 20-37 kg/m2. Subcutaneous abdominal adipose tissue biopsies were obtained and stromal cell populations identified by flow cytometry. Sorted cells underwent in vitro cultivation via traditional mesenchymal culture methodology (mesenchymal lineage) or a novel 3D-fibrin clot followed by traditional adherent culture (hematopoietic lineage) for assessment of proliferation and differentiation capacity. We found hematopoietic lineage cells isolated from the adipose tissue stroma, but not the circulation, were capable of proliferation and multilineage (adipogenic and osteogenic) differentiation in vitro. We provide a new investigative tool that can be used to perform translational studies of BMDAs and provide initial evidence that hematopoietic lineage cells isolated from the adipose tissue of humans can undergo hematopoietic-to-mesenchymal transition with multilineage differentiation potential in an in vitro environment.
Collapse
Affiliation(s)
- Kathleen M Gavin
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Bogdan Conrad
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Sullivan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Ruby Vianzon
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alistaire S Acosta
- University of Colorado Cancer Center Flow Cytometry Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wendy M Kohrt
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Dwight J Klemm
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| |
Collapse
|
12
|
Liu M, Lu F, Feng J. Aging and homeostasis of the hypodermis in the age-related deterioration of skin function. Cell Death Dis 2024; 15:443. [PMID: 38914551 PMCID: PMC11196735 DOI: 10.1038/s41419-024-06818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 02/01/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Adipose tissues in the hypodermis, the crucial stem cell reservoir in the skin and the endocrine organ for the maintenance of skin homeostasis undergo significant changes during skin aging. Dermal white adipose tissue (dWAT) has recently been recognized as an important organ for both non-metabolic and metabolic health in skin regeneration and rejuvenation. Defective differentiation, adipogenesis, improper adipocytokine production, and immunological dissonance dysfunction in dWAT lead to age-associated clinical changes. Here, we review age-related alterations in dWAT across levels, emphasizing the mechanisms underlying the regulation of aging. We also discuss the pathogenic changes involved in age-related fat dysfunction and the unfavorable consequences of accelerated skin aging, such as chronic inflammaging, immunosenescence, delayed wound healing, and fibrosis. Research has shown that adipose aging is an early initiation event and a potential target for extending longevity. We believe that adipose tissues play an essential role in aging and form a potential therapeutic target for the treatment of age-related skin diseases. Further research is needed to improve our understanding of this phenomenon.
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
13
|
Zhang Y, Zhang B, Sun X. The molecular mechanism of macrophage-adipocyte crosstalk in maintaining energy homeostasis. Front Immunol 2024; 15:1378202. [PMID: 38650945 PMCID: PMC11033412 DOI: 10.3389/fimmu.2024.1378202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Interactions between macrophages and adipocytes in adipose tissue are critical for the regulation of energy metabolism and obesity. Macrophage polarization induced by cold or other stimulations can drive metabolic reprogramming of adipocytes, browning, and thermogenesis. Accordingly, investigating the roles of macrophages and adipocytes in the maintenance of energy homeostasis is critical for the development of novel therapeutic approaches specifically targeting macrophages in metabolic disorders such as obesity. Current review outlines macrophage polarization not only regulates the release of central nervous system and inflammatory factors, but controls mitochondrial function, and other factor that induce metabolic reprogramming of adipocytes and maintain energy homeostasis. We also emphasized on how the adipocytes conversely motivate the polarization of macrophage. Exploring the interactions between adipocytes and macrophages may provide new therapeutic strategies for the management of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yudie Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Eisinger K, Girke P, Buechler C, Krautbauer S. Adipose tissue depot specific expression and regulation of fibrosis-related genes and proteins in experimental obesity. Mamm Genome 2024; 35:13-30. [PMID: 37884762 PMCID: PMC10884164 DOI: 10.1007/s00335-023-10022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Transforming growth factor beta (Tgfb) is a well-studied pro-fibrotic cytokine, which upregulates cellular communication network factor 2 (Ccn2), collagen, and actin alpha 2, smooth muscle (Acta2) expression. Obesity induces adipose tissue fibrosis, which contributes to metabolic diseases. This work aimed to analyze the expression of Tgfb, Ccn2, collagen1a1 (Col1a1), Acta2 and BMP and activin membrane-bound inhibitor (Bambi), which is a negative regulator of Tgfb signaling, in different adipose tissue depots of mice fed a standard chow, mice fed a high fat diet (HFD) and ob/ob mice. Principally, these genes were low expressed in brown adipose tissues and this difference was less evident for the ob/ob mice. Ccn2 and Bambi protein as well as mRNA expression, and collagen1a1 mRNA were not induced in the adipose tissues upon HFD feeding whereas Tgfb and Acta2 mRNA increased in the white fat depots. Immunoblot analysis showed that Acta2 protein was higher in subcutaneous and perirenal fat of these mice. In the ob/ob mice, Ccn2 mRNA and Ccn2 protein were upregulated in the fat depots. Here, Tgfb, Acta2 and Col1a1 mRNA levels and serum Tgfb protein were increased. Acta2 protein was, however, not higher in subcutaneous and perirenal fat of these mice. Col6a1 mRNA was shown before to be higher in obese fat tissues. Current analysis proved the Col6a1 protein was induced in subcutaneous fat of HFD fed mice. Notably, Col6a1 was reduced in perirenal fat of ob/ob mice in comparison to the respective controls. 3T3-L1 cells express Ccn2 and Bambi protein, whose levels were not changed by fatty acids, leptin, lipopolysaccharide, tumor necrosis factor and interleukin-6. All of these factors led to higher Tgfb in 3T3-L1 adipocyte media but did not increase its mRNA levels. Free fatty acids induced necrosis whereas apoptosis did not occur in any of the in vitro incubations excluding cell death as a main reason for higher Tgfb in cell media. In summary, Tgfb mRNA is consistently induced in white fat tissues in obesity but this is not paralleled by a clear increase of its target genes. Moreover, discrepancies between mRNA and protein expression of Acta2 were observed. Adipocytes seemingly do not contribute to higher Tgfb mRNA levels in obesity. These cells release more Tgfb protein when challenged with obesity-related metabolites connecting metabolic dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, 93040, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| | - Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| |
Collapse
|
15
|
Hu H, Zhang Z, Liu Z, Chu F, Ran J, Liang W. Thickened Perirenal Fat Predicts Poor Renal Outcome in Patients with Immunoglobulin A Nephropathy: A Population-Based Retrospective Cohort Study. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:51-60. [PMID: 38322631 PMCID: PMC10843190 DOI: 10.1159/000533507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 08/07/2023] [Indexed: 02/08/2024]
Abstract
Introduction Perirenal fat is a pad that fills the retroperitoneal space outside the kidney, which affects kidney function in various ways. However, the association between perirenal fat and IgA nephropathy (IgAN) has not yet been elucidated. This study aimed to investigate the role of perirenal fat in predicting IgAN progression. Methods A total of 473 patients with biopsy-proven IgAN and follow-up information were recruited, and perirenal fat thickness (PFT) was measured using color Doppler ultrasonography at renal biopsy. Patients were divided into two groups according to the median PFT: the low-PFT group (PFT ≤1.34 cm, n = 239) and the high PFT group (PFT >1.35 cm, n = 234). A total of 473 healthy participants were included in the control group. Basic clinical characteristics were assessed at the time of renal biopsy, and the relationship between PFT and combined endpoints was analyzed. The renal composite endpoints were defined as a two-fold increase in blood creatinine level, end-stage renal disease (dialysis over 3 months). Kaplan-Meier survival analysis was used to explore the role of PFT in the progression of IgAN. Three clinicopathological models of multivariate Cox regression analysis were established to evaluate the association between PFT and renal prognosis in patients with IgAN. Results Compared to healthy subjects, patients with IgAN showed significantly higher PFT. After a median follow-up of 50 months, 75 of 473 patients (15.9%) with IgAN reached renal composite endpoints. Among those, 13 of 239 patients (5.4%) were in the low PFT group, and 62 of 234 patients (26.5%) were in the high PFT group (p < 0.001). The results of three Cox regression models (including demographics, pathological and clinical indicators, and PFT) demonstrated that a higher PFT was significantly associated with a higher risk of reaching renal composite endpoints in patients with IgAN. Conclusion This study indicated a positive relationship between PFT at renal biopsy and renal progression in patients with IgAN, suggesting that perirenal fat might act as a marker of poor prognosis in patients with IgAN.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Clinical Research Center of Kidney Disease, Wuhan, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Clinical Research Center of Kidney Disease, Wuhan, China
| | - Zikang Liu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Clinical Research Center of Kidney Disease, Wuhan, China
| | - Fan Chu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Clinical Research Center of Kidney Disease, Wuhan, China
| | - Jialu Ran
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Clinical Research Center of Kidney Disease, Wuhan, China
| |
Collapse
|
16
|
Gyurina K, Yarmak M, Sasi-Szabó L, Molnár S, Méhes G, Röszer T. Loss of Uncoupling Protein 1 Expression in the Subcutaneous Adipose Tissue Predicts Childhood Obesity. Int J Mol Sci 2023; 24:16706. [PMID: 38069028 PMCID: PMC10706300 DOI: 10.3390/ijms242316706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Stimulation of thermogenesis by inducing uncoupling protein 1 (UCP1) expression in adipocytes is thought to promote weight loss by increasing energy expenditure, and it is postulated that the human newborn has thermogenic subcutaneous fat depots. However, it remains unclear whether a relevant number of UCP1-expressing (UCP1+) adipocytes exist in the early postnatal life. Here we studied the distribution of UCP1 and the expression of thermogenic genes in the subcutaneous adipose tissues of the human fetus, infant and child. We show that the deep layer of human fetal and neonatal subcutaneous fat, particularly the abdominal wall, is rich in UCP1+ adipocytes. These adipocytes develop in the late third trimester and persist throughout childhood, expressing a panel of genes linked to mitochondrial biogenesis and thermogenesis. During the early childhood adiposity rebound-a critical phase that determines obesity risk later in life-the absence of adipose tissue UCP1 expression in children with normal body mass index (BMI) correlates with an obesity-associated gene expression signature. Finally, UCP1 expression is negatively correlated with BMI z-score and adipocyte size in infants and children. Overall, our results show that the absence of UCP1 expression in adipose tissue is an early indicator of adipose tissue expansion in children.
Collapse
Affiliation(s)
- Katalin Gyurina
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Mariia Yarmak
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - László Sasi-Szabó
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Sarolta Molnár
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Tamás Röszer
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
- Institute of Neurobiology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
17
|
Li Z, Rosen CJ. The Multifaceted Roles of Bone Marrow Adipocytes in Bone and Hematopoietic Homeostasis. J Clin Endocrinol Metab 2023; 108:e1465-e1472. [PMID: 37315208 DOI: 10.1210/clinem/dgad355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023]
Abstract
Bone marrow adipose tissue (BMAT) makes up a significant portion of the marrow space, ranging from 50% to 70%, in healthy adults. It expands with aging, obesity, anorexia nervosa, and irradiation, which are conditions associated with skeletal complications or hematopoietic disorders. Therefore, BMAT has been viewed as a negative component of the bone marrow niche for decades, although the mechanisms and causative relationships have not been well-addressed. Of note, recent studies have revealed that BMAT is a multifaceted tissue that can serve as an energy reservoir to fuel osteoblasts and hematopoietic cells under stressful situations, and also acts as an endocrine/paracrine organ to suppress bone formation and support hematopoiesis at steady-state conditions. In this review, we summarize the uniqueness of BMAT, the complex findings of previous studies, and update our understanding of the physiological roles of BMAT in bone and hematopoietic metabolism based on a newly established bone marrow adipocyte-specific mouse model.
Collapse
Affiliation(s)
- Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA
| | - Clifford J Rosen
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA
| |
Collapse
|
18
|
Rao J, Djeffal Y, Chal J, Marchianò F, Wang CH, Al Tanoury Z, Gapon S, Mayeuf-Louchart A, Glass I, Sefton EM, Habermann B, Kardon G, Watt FM, Tseng YH, Pourquié O. Reconstructing human brown fat developmental trajectory in vitro. Dev Cell 2023; 58:2359-2375.e8. [PMID: 37647896 PMCID: PMC10873093 DOI: 10.1016/j.devcel.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/23/2022] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
Brown adipocytes (BAs) represent a specialized cell type that is able to uncouple nutrient catabolism from ATP generation to dissipate energy as heat. In humans, the brown fat tissue is composed of discrete depots found throughout the neck and trunk region. BAs originate from a precursor common to skeletal muscle, but their developmental trajectory remains poorly understood. Here, we used single-cell RNA sequencing to characterize the development of interscapular brown fat in mice. Our analysis identified a transient stage of BA differentiation characterized by the expression of the transcription factor GATA6. We show that recapitulating the sequence of signaling cues identified in mice can lead to efficient differentiation of BAs in vitro from human pluripotent stem cells. These precursors can in turn be efficiently converted into functional BAs that can respond to signals mimicking adrenergic stimuli by increasing their metabolism, resulting in heat production.
Collapse
Affiliation(s)
- Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yannis Djeffal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jerome Chal
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Fabio Marchianò
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Chih-Hao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Ian Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Bianca Habermann
- Aix-Marseille University, CNRS, IBDM, The Turing Center for Living Systems, 13009 Marseille, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Great Maze Pond, London SE1 9RT, UK
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
19
|
Nilaweera KN, Cotter PD. Can dietary proteins selectively reduce either the visceral or subcutaneous adipose tissues? Obes Rev 2023; 24:e13613. [PMID: 37548066 DOI: 10.1111/obr.13613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 06/22/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
There is a considerable appeal for interventions that can selectively reduce either the visceral or subcutaneous white adipose tissues in humans and other species because of their associated impact on outcomes related to metabolic health. Here, we reviewed the data related to the specificity of five interventions to affect the two depots in humans and rodents. The interventions relate to the use of dietary proteins, monounsaturated fatty acids, polyunsaturated fatty acids, calorie restriction, or bariatric surgery. The available data show that calorie restriction and bariatric surgery reduce both visceral and subcutaneous tissues, whereas there is no consistency in the effect of monounsaturated or polyunsaturated fatty acids. Dietary proteins, more specifically, whey proteins show efficacy to reduce one or both depots based on how the proteins interact with other macronutrients in the diet. We provide evidence that this specificity is related to changes in the composition and the functional potential of the gut microbiota and the resulting metabolites produced by these microorganisms. The effect of the sex of the host is also discussed. This knowledge may help to develop nutritional approaches to deplete either the visceral or subcutaneous adipose tissues and improve metabolic health in humans and other species.
Collapse
Affiliation(s)
- Kanishka N Nilaweera
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- VistaMilk Research Centre, Teagasc, Fermoy, County Cork, Ireland
| | - Paul D Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- VistaMilk Research Centre, Teagasc, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Luo M, Wang H, Zhang J, Yixi K, Shu S, Fu C, Zhong J, Peng W. IMF deposition ceRNA network analysis and functional study of HIF1a in yak. Front Vet Sci 2023; 10:1272238. [PMID: 37915947 PMCID: PMC10616239 DOI: 10.3389/fvets.2023.1272238] [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: 08/03/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
The concentration of intramuscular fat (IMF) is a crucial determinant of yak meat quality. However, the molecular mechanisms that regulate IMF in yak remain largely elusive. In our study, we conducted transcriptome sequencing on the longissimus dorsi muscle tissues of yaks with varying IMF contents. We then filtered differentially expressed genes (DEGs), microRNAs (DEMs), and long non-coding RNAs (DELs) to elucidate potential regulatory pathways of adipogenesis in yaks. Overall, our research sheds light on an array of potential mRNAs and noncoding RNAs implicated in IMF deposition and elaborates on the role of HIF1α in yaks. These findings contribute valuable insights that can serve as a guide for further research into the molecular mechanisms governing IMF deposition.
Collapse
Affiliation(s)
- Mengning Luo
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Jun Zhang
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Kangzhu Yixi
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Shi Shu
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Changqi Fu
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Wei Peng
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| |
Collapse
|
21
|
Hwang MS, Park J, Ham Y, Lee IH, Chun KH. Roles of Protein Post-Translational Modifications During Adipocyte Senescence. Int J Biol Sci 2023; 19:5245-5256. [PMID: 37928271 PMCID: PMC10620833 DOI: 10.7150/ijbs.86404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023] Open
Abstract
Adipocytes are adipose tissues that supply energy to the body through lipids. The two main types of adipocytes comprise white adipocytes (WAT) that store energy, and brown adipocytes (BAT), which generate heat by burning stored fat (thermogenesis). Emerging evidence indicates that dysregulated adipocyte senescence may disrupt metabolic homeostasis, leading to various diseases and aging. Adipocytes undergo senescence via irreversible cell-cycle arrest in response to DNA damage, oxidative stress, telomere dysfunction, or adipocyte over-expansion upon chronic lipid accumulation. The amount of detectable BAT decreases with age. Activation of cell cycle regulators and dysregulation of adipogenesis-regulating factors may constitute a molecular mechanism that accelerates adipocyte senescence. To better understand the regulation of adipocyte senescence, the effects of post-translational modifications (PTMs), is essential for clarifying the activity and stability of these proteins. PTMs are covalent enzymatic protein modifications introduced following protein biosynthesis, such as phosphorylation, acetylation, ubiquitination, or glycosylation. Determining the contribution of PTMs to adipocyte senescence may identify new therapeutic targets for the regulation of adipocyte senescence. In this review, we discuss a conceptual case in which PTMs regulate adipocyte senescence and explain the mechanisms underlying protein regulation, which may lead to the development of effective strategies to combat metabolic diseases.
Collapse
Affiliation(s)
- Min-Seon Hwang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jingyeong Park
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Yunha Ham
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - In Hye Lee
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
22
|
Palacios-Marin I, Serra D, Jimenez-Chillarón J, Herrero L, Todorčević M. Adipose Tissue Dynamics: Cellular and Lipid Turnover in Health and Disease. Nutrients 2023; 15:3968. [PMID: 37764752 PMCID: PMC10535304 DOI: 10.3390/nu15183968] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
The alarming increase in obesity and its related metabolic health complications, such as type 2 diabetes, has evolved into a global pandemic. Obesity is mainly characterized by excessive accumulation of adipose tissue, primarily due to an imbalance between energy intake and expenditure. Prolonged positive energy balance leads to the expansion of existing adipocytes (hypertrophy) and/or an increase in preadipocyte and adipocyte number (hyperplasia) to accommodate excess energy intake. However, obesity is not solely defined by increases in adipocyte size and number. The turnover of adipose tissue cells also plays a crucial role in the development and progression of obesity. Cell turnover encompasses the processes of cell proliferation, differentiation, and apoptosis, which collectively regulate the overall cell population within adipose tissue. Lipid turnover represents another critical factor that influences how adipose tissue stores and releases energy. Our understanding of adipose tissue lipid turnover in humans remains limited due to the slow rate of turnover and methodological constraints. Nonetheless, disturbances in lipid metabolism are strongly associated with altered adipose tissue lipid turnover. In obesity, there is a decreased rate of triglyceride removal (lipolysis followed by oxidation), leading to the accumulation of triglycerides over time. This review provides a comprehensive summary of findings from both in vitro and in vivo methods used to study the turnover of adipose cells and lipids in metabolic health and disease. Understanding the mechanisms underlying cellular and lipid turnover in obesity is essential for developing strategies to mitigate the adverse effects of excess adiposity.
Collapse
Affiliation(s)
- Ivonne Palacios-Marin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Josep Jimenez-Chillarón
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Physiological Sciences, School of Medicine, University of Barcelona, E-08907 L’Hospitalet, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Marijana Todorčević
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
23
|
Wess J, Oteng AB, Rivera-Gonzalez O, Gurevich EV, Gurevich VV. β-Arrestins: Structure, Function, Physiology, and Pharmacological Perspectives. Pharmacol Rev 2023; 75:854-884. [PMID: 37028945 PMCID: PMC10441628 DOI: 10.1124/pharmrev.121.000302] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
The two β-arrestins, β-arrestin-1 and -2 (systematic names: arrestin-2 and -3, respectively), are multifunctional intracellular proteins that regulate the activity of a very large number of cellular signaling pathways and physiologic functions. The two proteins were discovered for their ability to disrupt signaling via G protein-coupled receptors (GPCRs) via binding to the activated receptors. However, it is now well recognized that both β-arrestins can also act as direct modulators of numerous cellular processes via either GPCR-dependent or -independent mechanisms. Recent structural, biophysical, and biochemical studies have provided novel insights into how β-arrestins bind to activated GPCRs and downstream effector proteins. Studies with β-arrestin mutant mice have identified numerous physiologic and pathophysiological processes regulated by β-arrestin-1 and/or -2. Following a short summary of recent structural studies, this review primarily focuses on β-arrestin-regulated physiologic functions, with particular focus on the central nervous system and the roles of β-arrestins in carcinogenesis and key metabolic processes including the maintenance of glucose and energy homeostasis. This review also highlights potential therapeutic implications of these studies and discusses strategies that could prove useful for targeting specific β-arrestin-regulated signaling pathways for therapeutic purposes. SIGNIFICANCE STATEMENT: The two β-arrestins, structurally closely related intracellular proteins that are evolutionarily highly conserved, have emerged as multifunctional proteins able to regulate a vast array of cellular and physiological functions. The outcome of studies with β-arrestin mutant mice and cultured cells, complemented by novel insights into β-arrestin structure and function, should pave the way for the development of novel classes of therapeutically useful drugs capable of regulating specific β-arrestin functions.
Collapse
Affiliation(s)
- Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Antwi-Boasiako Oteng
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Osvaldo Rivera-Gonzalez
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Eugenia V Gurevich
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| | - Vsevolod V Gurevich
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland (J.W., A.-B.O., O.R.-G.); and Department of Pharmacology, Vanderbilt University, Nashville, Tennessee (E.V.G., V.V.G.)
| |
Collapse
|
24
|
Desevin K, Cortez BN, Lin JZ, Lama D, Layne MD, Farmer SR, Rabhi N. Adrenergic Reprogramming of Preexisting Adipogenic Trajectories Steer Naïve Mural Cells Toward Beige Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554950. [PMID: 37662295 PMCID: PMC10473761 DOI: 10.1101/2023.08.26.554950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
In adult white adipose tissue, cold or β3-adrenoceptor activation promotes the appearance of thermogenic beige adipocytes. Our comprehensive single-cell analysis revealed that these cells arise through the reprogramming of existing adipogenic trajectories, rather than from a single precursor. These trajectories predominantly arise from SM22-expressing vascular mural progenitor cells. Central in this transition is the activation of Adrb3 in mature adipocytes, leading to subsequent upregulation of Adrb1 in primed progenitors. Under thermoneutral conditions, synergistic activation of both Adrb3 and Adrb1 recapitulates the pattern of cold-induced SM22+ cell recruitment. Lipolysis-derived eicosanoids, specifically docosahexaenoic acid (DHA) and arachidonic acid (AA) prime these processes and in vitro, were sufficient to recapitulate progenitor cells priming. Collectively, our findings provide a robust model for cold-induced beige adipogenesis, emphasizing a profound relationship between mature adipocytes and mural cells during cold acclimation, and revealing the metabolic potential of this unique cellular reservoir.
Collapse
|
25
|
Souza-Tavares H, Miranda CS, Vasques-Monteiro IML, Sandoval C, Santana-Oliveira DA, Silva-Veiga FM, Fernandes-da-Silva A, Souza-Mello V. Peroxisome proliferator-activated receptors as targets to treat metabolic diseases: Focus on the adipose tissue, liver, and pancreas. World J Gastroenterol 2023; 29:4136-4155. [PMID: 37475842 PMCID: PMC10354577 DOI: 10.3748/wjg.v29.i26.4136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023] Open
Abstract
The world is experiencing reflections of the intersection of two pandemics: Obesity and coronavirus disease 2019. The prevalence of obesity has tripled since 1975 worldwide, representing substantial public health costs due to its comorbidities. The adipose tissue is the initial site of obesity impairments. During excessive energy intake, it undergoes hyperplasia and hypertrophy until overt inflammation and insulin resistance turn adipocytes into dysfunctional cells that send lipotoxic signals to other organs. The pancreas is one of the organs most affected by obesity. Once lipotoxicity becomes chronic, there is an increase in insulin secretion by pancreatic beta cells, a surrogate for type 2 diabetes mellitus (T2DM). These alterations threaten the survival of the pancreatic islets, which tend to become dysfunctional, reaching exhaustion in the long term. As for the liver, lipotoxicity favors lipogenesis and impairs beta-oxidation, resulting in hepatic steatosis. This silent disease affects around 30% of the worldwide population and can evolve into end-stage liver disease. Although therapy for hepatic steatosis remains to be defined, peroxisome proliferator-activated receptors (PPARs) activation copes with T2DM management. Peroxisome PPARs are transcription factors found at the intersection of several metabolic pathways, leading to insulin resistance relief, improved thermogenesis, and expressive hepatic steatosis mitigation by increasing mitochondrial beta-oxidation. This review aimed to update the potential of PPAR agonists as targets to treat metabolic diseases, focusing on adipose tissue plasticity and hepatic and pancreatic remodeling.
Collapse
Affiliation(s)
| | | | | | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Osorno 5310431, Chile
- Departamento de Ciencias Preclínicas, Universidad de la Frontera, Temuco 4780000, Chile
| | | | | | | | - Vanessa Souza-Mello
- Department of Anatomy, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| |
Collapse
|
26
|
Yuan Y, Fan Y, Zhou Y, Qiu R, Kang W, Liu Y, Chen Y, Wang C, Shi J, Liu C, Li Y, Wu M, Huang K, Liu Y, Zheng L. Linker histone variant H1.2 is a brake on white adipose tissue browning. Nat Commun 2023; 14:3982. [PMID: 37414781 PMCID: PMC10325996 DOI: 10.1038/s41467-023-39713-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
Adipose-tissue is a central metabolic organ for whole-body energy homeostasis. Here, we find that highly expressed H1.2, a linker histone variant, senses thermogenic stimuli in beige and brown adipocytes. Adipocyte H1.2 regulates thermogenic genes in inguinal white-adipose-tissue (iWAT) and affects energy expenditure. Adipocyte H1.2 deletion (H1.2AKO) male mice show promoted iWAT browning and improved cold tolerance; while overexpressing H1.2 shows opposite effects. Mechanistically, H1.2 binds to the promoter of Il10rα, which encodes an Il10 receptor, and positively regulates its expression to suppress thermogenesis in a beige cell autonomous manner. Il10rα overexpression in iWAT negates cold-enhanced browning of H1.2AKO male mice. Increased H1.2 level is also found in WAT of obese humans and male mice. H1.2AKO male mice show alleviated fat accumulation and glucose intolerance in long-term normal chow-fed and high fat diet-fed conditions; while Il10rα overexpression abolishes these effects. Here, we show a metabolic function of H1.2-Il10rα axis in iWAT.
Collapse
Affiliation(s)
- Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Yu Fan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Yihao Zhou
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Rong Qiu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Wei Kang
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Yu Liu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Yuchen Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Chenyu Wang
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Jiajian Shi
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yangkai Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Min Wu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, 430072, Wuhan, China.
| |
Collapse
|
27
|
Ricci M, De Feo MS, Granese GM, Frantellizzi V, Carabellese B, Lubrano E, Cimini A. 18F-FDG PET/CT technology for the assessment of brown adipose tissue: an updated review. Expert Rev Med Devices 2023; 20:1143-1156. [PMID: 37965719 DOI: 10.1080/17434440.2023.2283618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/10/2023] [Indexed: 11/16/2023]
Abstract
INTRODUCTION This review provides an update of 18 F-fluorodeoxyglucose ([18F] FDG) for Brown adipose tissue (BAT) activity quantification, whose role is not completely understood. AREAS COVERED We conducted an unstructured search of the literature for any studies employing the [18F] FDG PET in BAT assessment. We explored BAT quantification both in healthy individuals and in different pathologies, after cold exposure and as a metabolic biomarker. The assessment of possible BAT modulators by using [18F] FDG PET is shown. Further PET tracers and novel developments for BAT assessments are also described. EXPERT OPINION Further PET tracers and imaging modalities are under investigation, but the [18F] FDG PET is currently the method of choice for the evaluation of BAT and further multicentric trials are needed for a better understanding of the BAT physiopathology, also after cold stimuli. The modulation of BAT activity, assessed by [18F] FDG PET imaging, seems a promising tool for the management of conditions such as obesity and type 2 diabetes. Moreover, an interesting possible correlation of BAT activation with prognostic [18F] FDG PET indices in cancer patients should be assessed with further multicentric trials.
Collapse
Affiliation(s)
- Maria Ricci
- Nuclear Medicine Unit, Cardarelli Hospital, Campobasso, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences Oncology and Anatomo-Pathology, Sapienza University of Rome, Rome, Italy
| | - Giorgia Maria Granese
- Department of Radiological Sciences Oncology and Anatomo-Pathology, Sapienza University of Rome, Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences Oncology and Anatomo-Pathology, Sapienza University of Rome, Rome, Italy
| | | | - Ennio Lubrano
- Dipartimento di Medicina e Scienze della Salute, Università degli Studi del Molise, Italy
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Andrea Cimini
- Nuclear Medicine Unit, St. Salvatore Hospital, L'Aquila, Italy
| |
Collapse
|
28
|
Saavedra-Peña RDM, Taylor N, Flannery C, Rodeheffer MS. Estradiol cycling drives female obesogenic adipocyte hyperplasia. Cell Rep 2023; 42:112390. [PMID: 37053070 PMCID: PMC10567995 DOI: 10.1016/j.celrep.2023.112390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/21/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
White adipose tissue (WAT) distribution is sex dependent. Adipocyte hyperplasia contributes to WAT distribution in mice driven by cues in the tissue microenvironment, with females displaying hyperplasia in subcutaneous and visceral WAT, while males and ovariectomized females have visceral WAT (VWAT)-specific hyperplasia. However, the mechanism underlying sex-specific hyperplasia remains elusive. Here, transcriptome analysis in female mice shows that high-fat diet (HFD) induces estrogen signaling in adipocyte precursor cells (APCs). Analysis of APCs throughout the estrous cycle demonstrates increased proliferation only when proestrus (high estrogen) coincides with the onset of HFD feeding. We further show that estrogen receptor α (ERα) is required for this proliferation and that estradiol treatment at the onset of HFD feeding is sufficient to drive it. This estrous influence on APC proliferation leads to increased obesity driven by adipocyte hyperplasia. These data indicate that estrogen drives ERα-dependent obesogenic adipocyte hyperplasia in females, exacerbating obesity and contributing to the differential fat distribution between the sexes.
Collapse
Affiliation(s)
- Rocío Del M Saavedra-Peña
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Natalia Taylor
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Clare Flannery
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT 06520, USA; Section of Endocrinology and Metabolism, Yale University, New Haven, CT 06520, USA
| | - Matthew S Rodeheffer
- Department of Comparative Medicine, Yale University, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA; Yale Center for Molecular and Systems Metabolism, Yale University, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Puche-Juarez M, Toledano JM, Ochoa JJ, Diaz-Castro J, Moreno-Fernandez J. Influence of Adipose Tissue on Early Metabolic Programming: Conditioning Factors and Early Screening. Diagnostics (Basel) 2023; 13:diagnostics13091510. [PMID: 37174902 PMCID: PMC10177621 DOI: 10.3390/diagnostics13091510] [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: 03/08/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Obesity and being overweight have become one of the world's most severe health issues, not only because of the pathology but also because of the development of related comorbidities. Even when children reach adulthood, the mother's environment during pregnancy has been found to have a significant impact on obesity prevention in children. Thus, both maternal dietary habits and other factors such as gestational diabetes mellitus, excessive weight gain during pregnancy, smoking, or endocrine factors, among others, could influence newborn growth, adiposity, and body composition at birth, in childhood and adolescence, hence programming health in adulthood. METHODS The aim of this review is to analyze the most recent human studies on the programming of fetal adipose tissue to determine which modifiable factors may influence adiposity and thus prevent specific disorders later in life by means of a bibliographic review of articles related to the subject over the last ten years. CONCLUSIONS The importance of a healthy diet and lifestyle not only during pregnancy and the first months of life but also throughout childhood, especially during the first two years of life as this is a period of great plasticity, where the foundations for optimal health in later life will be laid, preventing the emergence of noncommunicable diseases including obesity, diabetes mellitus type 2, hypertension, being overweight, and any other pathology linked to metabolic syndrome, which is so prevalent today, through health programs beginning at a young age.
Collapse
Affiliation(s)
- Maria Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, E-18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, E-18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, E-18071 Granada, Spain
| | - Juan M Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, E-18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, E-18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, E-18071 Granada, Spain
| | - Julio J Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, E-18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, E-18071 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, E-18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, E-18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), E-18016 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, E-18071 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, E-18071 Granada, Spain
| |
Collapse
|
30
|
Zhang W, Raza SHA, Li B, Sun B, Wang S, Pant SD, Al-Abbas NS, Shaer NA, Zan L. miR-33a Inhibits the Differentiation of Bovine Preadipocytes through the IRS2-Akt Pathway. Genes (Basel) 2023; 14:529. [PMID: 36833456 PMCID: PMC9957011 DOI: 10.3390/genes14020529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/09/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Several microRNAs (miRNAs) are known to participate in adipogenesis. However, their role in this process, especially in the differentiation of bovine preadipocytes, remains to be elucidated. This study was intended to clarify the effect of microRNA-33a (miR-33a) on the differentiation of bovine preadipocytes by cell culture, real-time fluorescent quantitative PCR (qPCR), Oil Red staining, BODIPY staining, and Western blotting. The results indicate that overexpression of miR-33a significantly inhibited lipid droplet accumulation and decreased the mRNA and protein expression of adipocyte differentiation marker genes such as peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element-binding protein 1 (SREBP1), and fatty acid-binding protein 4 (FABP4). In contrast, the interference expression of miR-33a promoted lipid droplet accumulation and increased the expression of marker genes. Additionally, miR-33a directly targeted insulin receptor substrate 2 (IRS2) and regulated the phosphorylation level of serine/threonine kinase (Akt). Furthermore, miR-33a inhibition could rescue defects in the differentiation of bovine preadipocytes and the Akt phosphorylation level caused by small interfering IRS2 (si-IRS2). Collectively, these results indicate that miR-33a could inhibit the differentiation of bovine preadipocytes, possibly through the IRS2-Akt pathway. These findings might help develop practical means to improve the quality of beef.
Collapse
Affiliation(s)
- Wenzhen Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Safety of Livestock and Poultry Products, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Bingzhi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Bing Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Sihu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Sameer D. Pant
- Gulbali Institute, Charles Sturt University, Boorooma Street, Wagga Wagga, NSW 2678, Australia
| | - Nouf S. Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Nehad A. Shaer
- Department of Chemistry, Al Lieth University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
31
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
32
|
Chu T, Yang MS. A Review of Structural Features, Biological Functions and Biotransformation Studies in Adipose Tissues and an Assessment of Progress and Implications. Endocr Metab Immune Disord Drug Targets 2023; 23:12-20. [PMID: 36043732 DOI: 10.2174/1871530322666220827145241] [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: 03/18/2022] [Revised: 06/28/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022]
Abstract
Roles for adipose tissues in energy metabolism, health maintenance and disease onset have been established. Evidence indicates that white, brown and beige fats are quite different in terms of their cellular origin and biological characteristics. These differences are significant in targeting adipocytes to study the pathogenesis and prevention strategies of related diseases. The biotransformations of white, brown and beige fat cells constitute an intriguing topic worthy of further study, and the molecular mechanisms underlying the biotransformations of white, brown and beige fat cells remain to be elucidated. Hence, we herein collected evidence from studies on adipose tissue or adipocytes, and we extracted the structural features, biologic functions, and biotransformations of adipose tissue/adipocytes. The present review aimed to summarize the latest research progress and propose novel research directions with respect to adipose tissue and adipocytes. We posit that this work will provide new insights and opportunities in the effective treatment strategies for obesity, diabetes and other lipid-related diseases. It will also contribute to our knowledge of the basic biologic underpinnings of adipocyte biology.
Collapse
Affiliation(s)
- Ting Chu
- Department of Nursing, School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, People's Republic of China
| | - Mao Sheng Yang
- Laboratory of Disorders Genes and Department of Pharmacology, Jishou University School of Pharmacy, Jishou 416000, Hunan Province, People's Republic of China
| |
Collapse
|
33
|
Scamfer SR, Lee MD, Hilgendorf KI. Ciliary control of adipocyte progenitor cell fate regulates energy storage. Front Cell Dev Biol 2022; 10:1083372. [PMID: 36561368 PMCID: PMC9763467 DOI: 10.3389/fcell.2022.1083372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The primary cilium is a cellular sensory organelle found in most cells in our body. This includes adipocyte progenitor cells in our adipose tissue, a complex organ involved in energy storage, endocrine signaling, and thermogenesis. Numerous studies have shown that the primary cilium plays a critical role in directing the cell fate of adipocyte progenitor cells in multiple adipose tissue types. Accordingly, diseases with dysfunctional cilia called ciliopathies have a broad range of clinical manifestations, including obesity and diabetes. This review summarizes our current understanding of how the primary cilium regulates adipocyte progenitor cell fate in multiple contexts and illustrates the importance of the primary cilium in regulating energy storage and adipose tissue function.
Collapse
Affiliation(s)
| | | | - Keren I. Hilgendorf
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
34
|
Hoang AC, Sasi-Szabó L, Pál T, Szabó T, Diedrich V, Herwig A, Landgraf K, Körner A, Röszer T. Mitochondrial RNA stimulates beige adipocyte development in young mice. Nat Metab 2022; 4:1684-1696. [PMID: 36443525 PMCID: PMC9771821 DOI: 10.1038/s42255-022-00683-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/10/2022] [Indexed: 11/30/2022]
Abstract
Childhood obesity is a serious public health crisis and a critical factor that determines future obesity prevalence. Signals affecting adipocyte development in early postnatal life have a strong potential to trigger childhood obesity; however, these signals are still poorly understood. We show here that mitochondrial (mt)RNA efflux stimulates transcription of nuclear-encoded genes for mitobiogenesis and thermogenesis in adipocytes of young mice and human infants. While cytosolic mtRNA is a potential trigger of the interferon (IFN) response, young adipocytes lack such a response to cytosolic mtRNA due to the suppression of IFN regulatory factor (IRF)7 expression by vitamin D receptor signalling. Adult and obese adipocytes, however, strongly express IRF7 and mount an IFN response to cytosolic mtRNA. In turn, suppressing IRF7 expression in adult adipocytes restores mtRNA-induced mitobiogenesis and thermogenesis and eventually mitigates obesity. Retrograde mitochondrion-to-nucleus signalling by mtRNA is thus a mechanism to evoke thermogenic potential during early adipocyte development and to protect against obesity.
Collapse
Affiliation(s)
| | - László Sasi-Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Pál
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Annika Herwig
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, Ulm, Germany.
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
35
|
Röszer T. Metabolic impact of adipose tissue macrophages in the early postnatal life. J Leukoc Biol 2022; 112:1515-1524. [PMID: 35899927 PMCID: PMC9796690 DOI: 10.1002/jlb.3mr0722-201r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/02/2022] [Indexed: 01/07/2023] Open
Abstract
Adipose tissue macrophages (ATMs) play key roles in metabolic inflammation, insulin resistance, adipose tissue fibrosis, and immune disorders associated with obesity. Research on ATM biology has mostly been conducted in the setting of adult obesity, since adipocyte hypertrophy is associated with a significant increase in ATM number. Signals that control ATM activation toward a proinflammatory or a proresolving phenotype also determine the developmental program and lipid metabolism of adipocytes after birth. ATMs are present at birth and actively participate in the synthesis of mediators, which induce lipolysis, mitobiogenesis, and mitochondrial uncoupling in adipocytes. ATMs in the newborn and the infant promote a lipolytic and fatty acid oxidizing adipocyte phenotype, which is essential to support the lipid-fueled metabolism, to maintain nonshivering thermogenesis and counteract an excessive adipose tissue expansion. Since adipose tissue metabolism in the early postnatal life determines obesity status in adulthood, early-life ATM functions may have a life-long impact.
Collapse
Affiliation(s)
- Tamás Röszer
- Division of Pediatric Obesity, Children's Hospital and Institute of PediatricsUniversity of DebrecenDebrecenHungary,Institute of NeurobiologyUlm UniversityUlmGermany
| |
Collapse
|
36
|
Xie T, Chen X, Liu C, Cai X, Xiang M, Liu S, Li R, Lin Z, Liu D, Dong M, Chen X, Zou M, Qiao P. New insight into the role of lipid metabolism-related proteins in rheumatic heart valve disease. Lipids Health Dis 2022; 21:110. [PMID: 36307855 DOI: 10.1186/s12944-022-01722-x] [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: 06/27/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE The aim of this study was to determine the expression of lipid metabolism-related proteins in rheumatic heart valve disease (RHVD). METHODS This retrospective study involved a total of 20 cases of moderate or severe rheumatic mitral valve stenosis and 4 cases of mitral regurgitation due to secondary causes from September 2018 to September 2021. The patients enrolled included 12 males and 12 females who underwent surgical excision of the mitral valve at the cardiac surgery department of Hainan General Hospital. The samples of mitral valve were collected during surgery treatment as the study group, and mitral valves collected from patients with ischemic heart disease were allocated into the control group. Hematoxylin-eosin (HE), oil red staining and immunohistochemical (IHC) staining were conducted to compare the expression of lipid metabolism-related proteins (ATP-binding cassette transporter A1 and acyl-coenzyme A: cholesterol acyltransferase-1), and real-time polymerase chain reaction (RT-PCR) was applied to compare the mRNA levels of ABCA1, ACAT1, and the inflammatory cytokines TNF-α, IL-10, and MCP-1. RESULTS In general, the rheumatic mitral valve showed leaflet thickening along with border adhesions and visible yellow fats. Oil red O staining also revealed the abovementioned results as well as fat cells. Both ABCA1 and ACAT1 were expressed in the rheumatic mitral valve via IHC, whereas only ACAT1 showed a faint level of expression in the ischemic mitral valve with no expression of ABCA1. In addition, compared with the ischemic mitral valve, RT-PCT showed increased mRNA expression levels of ABCA1, ACAT1, and the inflammatory cytokines TNF-α, IL-10, and MCP-1 (P < 0.05). After dividing the RMs into two groups for RT-PCR, we found that the higher the expression of ABCA1 and ACAT1 was, the lower the relative expression of inflammatory factors. CONCLUSION This study showed that adipose tissue, adipose cells, and lipid transport-related proteins were expressed strongly in the rheumatic mitral valve, suggesting that adipose tissue formation might be one of the important pathways in the pathology of rheumatic heart disease. In addition, adipose tissue and adipocytes were also involved in the inflammatory process. These data provide new insight into pathological mechanisms in rheumatic heart disease.
Collapse
Affiliation(s)
- Ting Xie
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China.
| | - Xuan Chen
- International College of Nursing, Hainan Vocational University of Science and Technology, Haikou, Hainan, China
| | - Cong Liu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingjiu Cai
- Department of Cardiovascular Medicine, Hainan General Hospital, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Mei Xiang
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Shiwu Liu
- Department of Cardiovascular Medicine, Hainan General Hospital, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Ruzheng Li
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Zhichuan Lin
- Department of Neurology, Hainan General Hospital, Haikou, Hainan, China
| | - Debing Liu
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Ming Dong
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China
| | - Xinzhong Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Minghui Zou
- Department of Cardiovascular Surgery, Guangzhou Women & Children's Medical Center, Guangzhou, Guangdong, China
| | - Ping Qiao
- Department of Cardiovascular Medicine, Hainan General Hospital, No.19 Xiuhua Road, Xiuying District, Haikou, 571000, Hainan, China.
| |
Collapse
|
37
|
Chechekhin VI, Kulebyakin KY, Kokaev RI, Tyurin-Kuzmin PA. GPCRs in the regulation of the functional activity of multipotent mesenchymal stromal cells. Front Cell Dev Biol 2022; 10:953374. [PMID: 36046341 PMCID: PMC9421028 DOI: 10.3389/fcell.2022.953374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022] Open
Abstract
Adipose tissue is one of the tissues in the human body that is renewed during the whole life. Dysregulation of this process leads to conditions such as obesity, metabolic syndrome, and type 2 diabetes. The key role in maintaining the healthy state of adipose tissue is played by a specific group of postnatal stem cells called multipotent mesenchymal stromal cells (MSCs). They are both precursors for new adipocytes and key paracrine regulators of adipose tissue homeostasis. The activity of MSCs is tightly adjusted to the needs of the organism. To ensure such coordination, MSCs are put under strict regulation which is realized through a wide variety of signaling mechanisms. They control aspects of MSC activity such as proliferation, differentiation, and production of signal molecules via alteration of MSC sensitivity to hormonal stimuli. In this regard, MSCs use all the main mechanisms of hormonal sensitivity regulation observed in differentiated cells, but at the same time, several unique regulatory mechanisms have been found in MSCs. In the presented review, we will cover these unique mechanisms as well as specifics of common mechanisms of regulation of hormonal sensitivity in stem cells.
Collapse
Affiliation(s)
- Vadim I. Chechekhin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin Yu. Kulebyakin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Romesh I. Kokaev
- Institute of Biomedical Investigations, The Affiliate of Vladikavkaz Scientific Centre of Russian Academy of Sciences, Vladikavkaz, Russia
| | - Pyotr A. Tyurin-Kuzmin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- *Correspondence: Pyotr A. Tyurin-Kuzmin,
| |
Collapse
|
38
|
Salehidoost R, Korbonits M. Glucose and lipid metabolism abnormalities in Cushing's syndrome. J Neuroendocrinol 2022; 34:e13143. [PMID: 35980242 DOI: 10.1111/jne.13143] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022]
Abstract
Prolonged excess of glucocorticoids (GCs) has adverse systemic effects leading to significant morbidities and an increase in mortality. Metabolic alterations associated with the high level of the GCs are key risk factors for the poor outcome. These include GCs causing excess gluconeogenesis via upregulation of key enzymes in the liver, a reduction of insulin sensitivity in skeletal muscle, liver and adipose tissue by inhibiting the insulin receptor signalling pathway, and inhibition of insulin secretion in beta cells leading to dysregulated glucose metabolism. In addition, chronic GC exposure leads to an increase in visceral adipose tissue, as well as an increase in lipolysis resulting in higher circulating free fatty acid levels and in ectopic fat deposition. Remission of hypercortisolism improves these metabolic changes, but very often does not result in full resolution of the abnormalities. Therefore, long-term monitoring of metabolic variables is needed even after the resolution of the excess GC levels.
Collapse
Affiliation(s)
- Rezvan Salehidoost
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
39
|
Lecoutre S, Lambert M, Drygalski K, Dugail I, Maqdasy S, Hautefeuille M, Clément K. Importance of the Microenvironment and Mechanosensing in Adipose Tissue Biology. Cells 2022; 11:cells11152310. [PMID: 35954152 PMCID: PMC9367348 DOI: 10.3390/cells11152310] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue is an adaptive mechanism that increases nutrient buffering capacity in response to an overall positive energy balance. Over the course of expansion, the adipose microenvironment undergoes continual remodeling to maintain its structural and functional integrity. However, in the long run, adipose tissue remodeling, typically characterized by adipocyte hypertrophy, immune cells infiltration, fibrosis and changes in vascular architecture, generates mechanical stress on adipose cells. This mechanical stimulus is then transduced into a biochemical signal that alters adipose function through mechanotransduction. In this review, we describe the physical changes occurring during adipose tissue remodeling, and how they regulate adipose cell physiology and promote obesity-associated dysfunction in adipose tissue.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Mélanie Lambert
- Labex Inflamex, Université Sorbonne Paris Nord, INSERM, F-93000 Bobigny, France;
| | - Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), IBPS, Sorbonne Université, F-75005 Paris, France;
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, F-75013 Paris, France
- Correspondence: or
| |
Collapse
|
40
|
Triiodothyronine (T3) promotes brown fat hyperplasia via thyroid hormone receptor α mediated adipocyte progenitor cell proliferation. Nat Commun 2022; 13:3394. [PMID: 35697700 PMCID: PMC9192766 DOI: 10.1038/s41467-022-31154-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
The thyroid hormone (TH)-controlled recruitment process of brown adipose tissue (BAT) is not fully understood. Here, we show that long-term treatment of T3, the active form of TH, increases the recruitment of thermogenic capacity in interscapular BAT of male mice through hyperplasia by promoting the TH receptor α-mediated adipocyte progenitor cell proliferation. Our single-cell analysis reveals the heterogeneous nature and hierarchical trajectory within adipocyte progenitor cells of interscapular BAT. Further analyses suggest that T3 facilitates cell state transition from a more stem-like state towards a more committed adipogenic state and promotes cell cycle progression towards a mitotic state in adipocyte progenitor cells, through mechanisms involving the action of Myc on glycolysis. Our findings elucidate the mechanisms underlying the TH action in adipocyte progenitors residing in BAT and provide a framework for better understanding of the TH effects on hyperplastic growth and adaptive thermogenesis in BAT depot at a single-cell level. Thyroid hormone (TH) action regulates brown adipose tissue thermogenic capacity through incompletely understood mechanisms. Here the authors report that T3, the active form of TH, increases thermogenic capacity via thyroid hormone receptor α-mediated hyperplasia of brown adipose tissue adipocyte progenitor cells.
Collapse
|
41
|
Guo R, You X, Meng K, Sha R, Wang Z, Yuan N, Peng Q, Li Z, Xie Z, Chen R, Feng Y. Single-Cell RNA Sequencing Reveals Heterogeneity of Myf5-Derived Cells and Altered Myogenic Fate in the Absence of SRSF2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105775. [PMID: 35460187 PMCID: PMC9218650 DOI: 10.1002/advs.202105775] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/05/2022] [Indexed: 06/14/2023]
Abstract
Splicing factor SRSF2 acts as a critical regulator for cell survival, however, it remains unknown whether SRSF2 is involved in myoblast proliferation and myogenesis. Here, knockdown of SRSF2 in myoblasts causes high rates of apoptosis and defective differentiation. Combined conditional knockout and lineage tracing approaches show that Myf5-cre mice lacking SRSF2 die immediately at birth and exhibit a complete absence of mature myofibers. Mutant Myf5-derived cells (tdtomato-positive cells) are randomly scattered in the myogenic and non-myogenic regions, indicating loss of the community effect required for skeletal muscle differentiation. Single-cell RNA-sequencing reveals high heterogeneity of myf5-derived cells and non-myogenic cells are significantly increased at the expense of skeletal muscle cells in the absence of SRSF2, reflecting altered cell fate. SRSF2 is demonstrated to regulate the entry of Myf5 cells into the myogenic program and ensures their survival by preventing precocious differentiation and apoptosis. In summary, SRSF2 functions as an essential regulator for Myf5-derived cells to respond correctly to positional cues and to adopt their myogenic fate.
Collapse
Affiliation(s)
- Ruochen Guo
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Xue You
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Rula Sha
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhenzhen Wang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ningyang Yuan
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Qian Peng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhigang Li
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Zhiqin Xie
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Ruijiao Chen
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| | - Ying Feng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong ProvinceJining Medical UniversityJining272067P. R. China
| |
Collapse
|
42
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
43
|
Parathyroid hormone (PTH) regulation of metabolic homeostasis: An old dog teaches us new tricks. Mol Metab 2022; 60:101480. [PMID: 35338013 PMCID: PMC8980887 DOI: 10.1016/j.molmet.2022.101480] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/11/2022] [Accepted: 03/16/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Late in the nineteenth century, it was theorized that a circulating product produced by the parathyroid glands could negatively impact skeletal homeostasis. A century later, intermittent administration of that protein, namely parathyroid hormone (PTH), was approved by the FDA and EMA as the first anabolic agent to treat osteoporosis. Yet, several unanswered but important questions remain about the skeletal actions of PTH. SCOPE OF REVIEW Current research efforts have focused on improving the efficacy of PTH treatment by designing structural analogs and identifying other targets (e.g., the PTH or the calcium sensing receptor). A unique but only recently described aspect of PTH action is its regulation of cellular bioenergetics and metabolism, namely in bone and adipose tissue but also in other tissues. The current review aims to provide a brief background on PTH's previously described actions on bone and highlights how PTH regulates osteoblast bioenergetics, contributing to greater bone formation. It will also shed light on how PTH could alter metabolic homeostasis through its actions in other cells and tissues, thereby impacting the skeleton in a cell non-autonomous manner. MAJOR CONCLUSIONS PTH administration enhances bone formation by targeting the osteoblast through transcriptional changes in several pathways; the most prominent is via adenyl cyclase and PKA. PTH and its related protein, PTHrP, also induce glycolysis and fatty acid oxidation in bone cells and drive lipolysis and thermogenic programming in adipocytes; the latter may indirectly but positively influence skeletal metabolism. While much work remains, alterations in cellular metabolism may also provide a novel mechanism related to PTH's temporal actions. Thus, the bioenergetic impact of PTH can be considered another of the myriad anabolic effects of PTH on the skeleton. Just as importantly from a translational perspective, the non-skeletal metabolic effects may lead to a better understanding of whole-body homeostasis along with new and improved therapies to treat musculoskeletal conditions.
Collapse
|
44
|
Integrating adipocyte insulin signaling and metabolism in the multi-omics era. Trends Biochem Sci 2022; 47:531-546. [PMID: 35304047 DOI: 10.1016/j.tibs.2022.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 12/16/2022]
Abstract
Insulin stimulates glucose uptake into adipocytes via mTORC2/AKT signaling and GLUT4 translocation and directs glucose carbons into glycolysis, glycerol for TAG synthesis, and de novo lipogenesis. Adipocyte insulin resistance is an early indicator of type 2 diabetes in obesity, a worldwide health crisis. Thus, understanding the interplay between insulin signaling and central carbon metabolism pathways that maintains adipocyte function, blood glucose levels, and metabolic homeostasis is critical. While classically viewed through the lens of individual enzyme-substrate interactions, advances in mass spectrometry are beginning to illuminate adipocyte signaling and metabolic networks on an unprecedented scale, yet this is just the tip of the iceberg. Here, we review how 'omics approaches help to elucidate adipocyte insulin action in cellular time and space.
Collapse
|
45
|
Kong S, Huang X, Cao H, Bai Y, Che Q, Nie H, Su Z. Anti-obesity effects of galacto-oligosaccharides in obese rats. Eur J Pharmacol 2022; 917:174728. [PMID: 34965390 DOI: 10.1016/j.ejphar.2021.174728] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/29/2022]
Abstract
Galacto-oligosaccharides (GOS) are commonly used as prebiotic with a variety of known metabolic benefits; however, whether GOS plays a protective role in obesity remains unknown. Here, we demonstrate that GOS prevented obesity in a rat model of obesity induced by a high-fat diet. Our results showed that GOS effectively slowed weight gain in diet-induced obese rats without affecting energy intake. GOS significantly suppressed the hypertrophy and hyperplasia of white adipose tissue and markedly reduced the ratio of the fat/body. Consistently, GOS significantly improved serum total cholesterol, triglycerides, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol levels, indicating the weight loss activity of GOS. Interestingly, GOS also significantly increased the expression levels of browning proteins, including uncoupling protein 1, peroxisome proliferator-activated receptor-γ, peroxisome proliferator-activated receptor-γ coactivator 1α, and PR domain 16, in both white and brown adipose tissue. Furthermore, we found that GOS markedly increased the expression levels of liver X receptor α, peroxisome proliferation-activated receptor-α, low-density lipoprotein receptor, and cholesterol 7α-hydroxylase proteins in the liver of obese rats. Taken together, we concluded that GOS inhibits obesity by accelerating the browning of white fat cells and the thermogenesis of brown fat cells and that GOS improves host lipid homeostasis by promoting cholesterol catabolism.
Collapse
Affiliation(s)
- Shang Kong
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xingjun Huang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hua Cao
- Guangdong Cosmetics Engineering & Technology Research Center, School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou, 510663, China
| | - Hong Nie
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
46
|
Chernukha I, Fedulova L, Kotenkova E. White, beige and brown adipose tissue: structure, function, specific features and possibility formation and divergence in pigs. FOODS AND RAW MATERIALS 2022. [DOI: 10.21603/2308-4057-2022-1-10-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Introduction. Traditionally, mammalian adipose tissue is divided into white (white adipose tissue – WAT) and brown (brown adipose tissue – BAT). While the functions of WAT are well known as the triglyceride depot, the role of BAT in mammalian physiology has been under close investigation. The first description of the role of BAT in maintaining thermogenesis dates back to 1961. This article offers a review of structural and functional specificity of white, beige and brown adipose tissue.
Results and discussion. The differences and descriptions of adipocytes and their impact on the maintenance of the main functions of the mammalian body are described in this manuscript. In particular, thermogenesis, stress response, obesity, type II diabetes. In addition to WAT and BAT, an intermediate form was also detected in the body – beige fat (BeAT or Brite). The opposite opinions regarding the presence of three types of adipose tissue in the human and animal bodies are presented. Studies on the identification of uncoupling proteins 1 and 3 and their role in the transformation of white fat into beige/brown are considered. Basically, the data on the factors of endogenous and exogenous nature on their formation are given on the example of the human body.
Conclusion. With an abundance of publications on the keywords: “white, brown fat”, these studies, in the overwhelming majority, are devoted to the role of these fats in the formation of human thermogenesis, the assessment of the impact on obesity. Pigs have also been suggested to lack functional BAT, which is a major cause of neonatal death in the swine industry, therefore the focus on investigating role of different types of adipose tissue in pigs seems very promising in order to understand whether there is a compensating mechanism of thermogenesis.
Collapse
Affiliation(s)
- Irina Chernukha
- V.M. Gorbatov Federal Research Center for Food Systems of RAS
| | - Liliya Fedulova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS
| | - Elena Kotenkova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS
| |
Collapse
|
47
|
Adipogenesis of ear mesenchymal stem cells (EMSCs): adipose biomarker-based assessment of genetic variation, adipocyte function, and brown/brite differentiation. Mol Cell Biochem 2022; 477:1053-1063. [PMID: 34997885 DOI: 10.1007/s11010-021-04350-8] [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: 07/19/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
Ear mesenchymal stem cells (EMSCs) have been investigated to differentiate into adipocytes, chondrocytes, and muscle cells in vitro. However, the factors controlling adipogenesis of this stem cell population in vitro, function, and type of adipocytes raised from them are still unclear. Here we found that genetics have a modest effect on adipogenic capacity of EMSCs. Adipocytes differentiated from EMSCs have a potential function in lipid metabolism as indicated by expression of lipogenic genes and this function of EMSC adipocytes is regulated by genetics. EMSCs failed to be differentiated into brite/brown adipocytes due to their lack of a thermogenic program, but adipocytes raised from EMSCs showed a fate of white adipocytes. Overall, our data suggest that EMSCs differentiate into functional white adipocytes in vitro and this is genetic-dependent.
Collapse
|
48
|
Zuccarini M, Lambertucci C, Carluccio M, Giuliani P, Ronci M, Spinaci A, Volpini R, Ciccarelli R, Di Iorio P. Multipotent Stromal Cells from Subcutaneous Adipose Tissue of Normal Weight and Obese Subjects: Modulation of Their Adipogenic Differentiation by Adenosine A 1 Receptor Ligands. Cells 2021; 10:cells10123560. [PMID: 34944069 PMCID: PMC8700077 DOI: 10.3390/cells10123560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Adenosine A1 receptor (A1R) activation, stimulating lipogenesis and decreasing insulin resistance, could be useful for metabolic syndrome management in obese subjects. Since full A1R agonists induce harmful side-effects, while partial agonists show a better pharmacological profile, we investigated the influence of two derivatives of the full A1R agonist 2-chloro-N6-cyclopentyladenosine (CCPA), C1 and C2 behaving as A1R partial agonists in animal models, on the adipogenic differentiation of stromal/stem cells (ASCs) from human subcutaneous adipose tissue, which mainly contribute to increase fat mass in obesity. The ASCs from normal-weight subjects showed increased proliferation and A1R expression but reduced adipogenic differentiation compared to obese individual-derived ASCs. Cell exposure to CCPA, C1, C2 or DPCPX, an A1R antagonist, did not affect ASC proliferation, while mainly C2 and DPCPX significantly decreased adipogenic differentiation of both ASC types, reducing the activity of glycerol-3-phosphate dehydrogenase and the expression of PPARγ and FABP-4, all adipogenic markers, and phosphorylation of Akt in the phosphatidylinositol-3-kinase pathway, which plays a key-role in adipogenesis. While requiring confirmation in in vivo models, our results suggest that A1R partial agonists or antagonists, by limiting ASC differentiation into adipocytes and, thereby, fat mass expansion, could favor development/worsening of metabolic syndrome in obese subjects without a dietary control.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Catia Lambertucci
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
- Stem TeCh Group, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy;
| | - Andrea Spinaci
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Rosaria Volpini
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
- Correspondence:
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| |
Collapse
|
49
|
Comparative Transcriptome Profiling of Young and Old Brown Adipose Tissue Thermogenesis. Int J Mol Sci 2021; 22:ijms222313143. [PMID: 34884947 PMCID: PMC8658479 DOI: 10.3390/ijms222313143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
Brown adipose tissue (BAT) is a major site for uncoupling protein 1 (UCP1)-mediated non-shivering thermogenesis. BAT dissipates energy via heat generation to maintain the optimal body temperature and increases energy expenditure. These energetic processes in BAT use large amounts of glucose and fatty acid. Therefore, the thermogenesis of BAT may be harnessed to treat obesity and related diseases. In mice and humans, BAT levels decrease with aging, and the underlying mechanism is elusive. Here, we compared the transcriptomic profiles of both young and aged BAT in response to thermogenic stimuli. The profiles were extracted from the GEO database. Intriguingly, aging does not cause transcriptional changes in thermogenic genes but upregulates several pathways related to the immune response and downregulates metabolic pathways. Acute severe CE upregulates several pathways related to protein folding. Chronic mild CE upregulates metabolic pathways, especially related to carbohydrate metabolism. Our findings provide a better understanding of the effects of aging and metabolic responses to thermogenic stimuli in BAT at the transcriptome level.
Collapse
|
50
|
Li F, Jing J, Movahed M, Cui X, Cao Q, Wu R, Chen Z, Yu L, Pan Y, Shi H, Shi H, Xue B. Epigenetic interaction between UTX and DNMT1 regulates diet-induced myogenic remodeling in brown fat. Nat Commun 2021; 12:6838. [PMID: 34824202 PMCID: PMC8617140 DOI: 10.1038/s41467-021-27141-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/05/2021] [Indexed: 02/04/2023] Open
Abstract
Brown adipocytes share the same developmental origin with skeletal muscle. Here we find that a brown adipocyte-to-myocyte remodeling also exists in mature brown adipocytes, and is induced by prolonged high fat diet (HFD) feeding, leading to brown fat dysfunction. This process is regulated by the interaction of epigenetic pathways involving histone and DNA methylation. In mature brown adipocytes, the histone demethylase UTX maintains persistent demethylation of the repressive mark H3K27me3 at Prdm16 promoter, leading to high Prdm16 expression. PRDM16 then recruits DNA methyltransferase DNMT1 to Myod1 promoter, causing Myod1 promoter hypermethylation and suppressing its expression. The interaction between PRDM16 and DNMT1 coordinately serves to maintain brown adipocyte identity while repressing myogenic remodeling in mature brown adipocytes, thus promoting their active brown adipocyte thermogenic function. Suppressing this interaction by HFD feeding induces brown adipocyte-to-myocyte remodeling, which limits brown adipocyte thermogenic capacity and compromises diet-induced thermogenesis, leading to the development of obesity.
Collapse
Affiliation(s)
- Fenfen Li
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Jia Jing
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Miranda Movahed
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Xin Cui
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Qiang Cao
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Rui Wu
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Ziyue Chen
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA
| | - Liqing Yu
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Yi Pan
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA ,grid.458489.c0000 0001 0483 7922Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 P.R. China
| | - Huidong Shi
- grid.410427.40000 0001 2284 9329Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA ,grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Hang Shi
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Bingzhong Xue
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| |
Collapse
|