51
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [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: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
52
|
Lee MJC, Saner NJ, Ferri A, García-Domínguez E, Broatch JR, Bishop DJ. Delineating the contribution of ageing and physical activity to changes in mitochondrial characteristics across the lifespan. Mol Aspects Med 2024; 97:101272. [PMID: 38626488 DOI: 10.1016/j.mam.2024.101272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/18/2024]
Abstract
Ageing is associated with widespread physiological changes prominent within all tissues, including skeletal muscle and the brain, which lead to a decline in physical function. To tackle the growing health and economic burdens associated with an ageing population, the concept of healthy ageing has become a major research priority. Changes in skeletal muscle mitochondrial characteristics have been suggested to make an important contribution to the reductions in skeletal muscle function with age, and age-related changes in mitochondrial content, respiratory function, morphology, and mitochondrial DNA have previously been reported. However, not all studies report changes in mitochondrial characteristics with ageing, and there is increasing evidence to suggest that physical activity (or inactivity) throughout life is a confounding factor when interpreting age-associated changes. Given that physical activity is a potent stimulus for inducing beneficial adaptations to mitochondrial characteristics, delineating the influence of physical activity on the changes in skeletal muscle that occur with age is complicated. This review aims to summarise our current understanding and knowledge gaps regarding age-related changes to mitochondrial characteristics within skeletal muscle, as well as to provide some novel insights into brain mitochondria, and to propose avenues of future research and targeted interventions. Furthermore, where possible, we incorporate discussions of the modifying effects of physical activity, exercise, and training status, to purported age-related changes in mitochondrial characteristics.
Collapse
Affiliation(s)
- Matthew J-C Lee
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Nicholas J Saner
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Alessandra Ferri
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Esther García-Domínguez
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - James R Broatch
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - David J Bishop
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
53
|
Singh S, Kriti M, K.S. A, Sarma DK, Verma V, Nagpal R, Mohania D, Tiwari R, Kumar M. Deciphering the complex interplay of risk factors in type 2 diabetes mellitus: A comprehensive review. Metabol Open 2024; 22:100287. [PMID: 38818227 PMCID: PMC11137529 DOI: 10.1016/j.metop.2024.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
The complex and multidimensional landscape of type 2 diabetes mellitus (T2D) is a major global concern. Despite several years of extensive research, the precise underlying causes of T2D remain elusive, but evidence suggests that it is influenced by a myriad of interconnected risk factors such as epigenetics, genetics, gut microbiome, environmental factors, organelle stress, and dietary habits. The number of factors influencing the pathogenesis is increasing day by day which worsens the scenario; meanwhile, the interconnections shoot up the frame. By gaining deeper insights into the contributing factors, we may pave the way for the development of personalized medicine, which could unlock more precise and impactful treatment pathways for individuals with T2D. This review summarizes the state of knowledge about T2D pathogenesis, focusing on the interplay between various risk factors and their implications for future therapeutic strategies. Understanding these factors could lead to tailored treatments targeting specific risk factors and inform prevention efforts on a population level, ultimately improving outcomes for individuals with T2D and reducing its burden globally.
Collapse
Affiliation(s)
- Samradhi Singh
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Mona Kriti
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Anamika K.S.
- Christ Deemed to Be University Bangalore, Karnataka, India
| | - Devojit Kumar Sarma
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, College of Health & Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Dheeraj Mohania
- Dr. R. P. Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Rajnarayan Tiwari
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Manoj Kumar
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| |
Collapse
|
54
|
Swart DH, de Haan M, Stevens J, Henning RH, Adel S, van der Graaf AC, Ulu N, Touw DJ, Krenning G. Safety, tolerability and toxicokinetics of the novel mitochondrial drug SUL-138 administered orally to rat and minipig. Toxicol Rep 2024; 12:345-355. [PMID: 38560508 PMCID: PMC10981007 DOI: 10.1016/j.toxrep.2024.03.009] [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: 12/13/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Noncommunicable Chronic Diseases (NCD) are a socioeconomic burden and considered one of the major health challenges for coming decades. Mitochondrial dysfunction has been implicated mechanistically in their pathophysiology. Therefore, targeting mitochondria holds great promise to improve clinical outcomes in NCDs. SUL-138, an orally bioavailable small molecule efficacious from 0.5 mg/kg, improves mitochondrial function during disease in several preclinical animal models. As preparation for a First-in-Human (FIH) trial, SUL-138 was investigated in 30-day GLP repeated dose toxicity studies in rat and minipig, selected based on their comparability with human metabolism, to determine toxicokinetics, potential toxicity and its reversibility. Rats were allocated to either vehicle, 27, 136 or 682 mg/kg SUL-138 dose groups and minipigs were allocated to either vehicle, 16, 82 or 409 mg/kg. Treatment occurred orally for 30 days followed by a recovery period of 14 days. During these studies clinical observations, toxicokinetic, clinical pathology, necropsy and histopathology evaluations were performed. There was significant systemic exposure to SUL-138 and toxicokinetics was characterized by a rapid absorption and elimination. In the rat, toxicokinetics was dose-proportional and AUC0-tlast ratios in both species indicated that SUL-138 does not accumulate in vivo. No treatment-related adverse effects were observed for dose levels up to 136 and 82 mg/kg/day in rat and minipig respectively. In conclusion, these preclinical studies demonstrate that SUL-138 is well tolerated after repeated administration in rat and minipig, with NOAELs of 136 and 82 mg/kg/day, respectively.
Collapse
Affiliation(s)
- Daniël H. Swart
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
- Sulfateq B.V., Admiraal de Ruyterlaan 5, Groningen 9726GN, the Netherlands
| | - Martin de Haan
- Sulfateq B.V., Admiraal de Ruyterlaan 5, Groningen 9726GN, the Netherlands
- Madeha Management & Consultancy B.V., Eilandseweg 10, Nederhorst den Berg 1394JE, the Netherlands
| | - Jasper Stevens
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Rob H. Henning
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Sovan Adel
- Sulfateq B.V., Admiraal de Ruyterlaan 5, Groningen 9726GN, the Netherlands
| | | | - Nadir Ulu
- Gen İlaç ve Sağlık Ürünleri A.Ş., Mustafa Kemal Mahallesi, 2119.Cad. No:3, Çankaya, Ankara 06520, Turkey
| | - Daan J. Touw
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen 9713GZ, the Netherlands
| | - Guido Krenning
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
- Sulfateq B.V., Admiraal de Ruyterlaan 5, Groningen 9726GN, the Netherlands
| |
Collapse
|
55
|
Gonzalez-Samano M, Villarreal HJ. Diabetes, life course and childhood socioeconomic conditions: an empirical assessment for Mexico. BMC Public Health 2024; 24:1274. [PMID: 38724956 PMCID: PMC11084140 DOI: 10.1186/s12889-024-18767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Demographic and epidemiological dynamics characterized by lower fertility rates and longer life expectancy, as well as higher prevalence of non-communicable diseases such as diabetes, represent important challenges for policy makers around the World. We investigate the risk factors that influence the diagnosis of diabetes in the Mexican population aged 50 years and over, including childhood poverty. RESULTS This work employs a probabilistic regression model with information from the Mexican Health and Aging Study (MHAS) of 2012 and 2018. Our results are consistent with the existing literature and should raise strong concerns. The findings suggest that risk factors that favor the diagnosis of diabetes in adulthood are: age, family antecedents of diabetes, obesity, and socioeconomic conditions during both adulthood and childhood. CONCLUSIONS Poverty conditions before the age 10, with inter-temporal poverty implications, are associated with a higher probability of being diagnosed with diabetes when older and pose extraordinary policy challenges.
Collapse
Affiliation(s)
- Marina Gonzalez-Samano
- Tecnologico de Monterrey, School of Government and Public Transformation, EGyTP, Mexico City, Mexico.
| | - Hector J Villarreal
- Tecnologico de Monterrey, School of Government and Public Transformation, EGyTP, Mexico City, Mexico
| |
Collapse
|
56
|
Mercier HW, Solinsky R, Taylor JA. Relationship of cardiometabolic disease risk factors with age and spinal cord injury duration. J Spinal Cord Med 2024; 47:379-386. [PMID: 35485952 PMCID: PMC11044727 DOI: 10.1080/10790268.2022.2065410] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
CONTEXT/OBJECTIVES Cardiometabolic disease (CMD) is increased after spinal cord injury (SCI), with an increased number of CMD risk factors that relate to higher mortality. The study objective was to characterize the relationship of age and injury duration with CMD. DESIGN Retrospective cohort assessment of CMD risks using unbiased recursive partitioning to divide for group comparison: (1) Lowest Risk, (2) Moderate Risk, and (3) Highest Risk based on classification and regression trees predicting CMD diagnosis by age and injury duration. SETTING Academic rehabilitation center laboratory. PARTICIPANTS Adults (N = 103; aged 18-75) with traumatic SCI (C4-L2) of 3 months to 42 years duration. INTERVENTIONS NA. OUTCOME MEASURES CMD risk factors (obesity, insulin resistance, dyslipidemia, and hypertension) using Paralyzed Veterans of America SCI-specific guidelines. RESULTS Obesity was prevalent (82%) and co-occurred with most other risk factors present. Age increased odds for CMD diagnosis by 1.05 per year (P = 0.02) and was directly related to elevated body mass index (BMI, β = 0.42, P < 0.05), fasting glucose (β = 0.58, P < 0.01), and higher systolic blood pressure (β = 0.31, P < 0.10). In contrast, time since injury contributed to lower risk factor count (β = -0.29, P < 0.10) and higher HDL-C (β = 0.50, P < 0.01), and was not related to odds of CMD diagnosis. CONCLUSION While SCI is linked to an increased risk of CMD, age is associated with higher CMD risk. Increased SCI duration related to improvement in individual CMD risk factors but did not decrease overall risk for CMD diagnosis. SCI may not uniformly increase CMD risks and highlight a necessary focus on weight management for risk prevention.
Collapse
Affiliation(s)
- Hannah W. Mercier
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Laboratory, Spaulding Rehabilitation Hospital, Cambridge, Massachusetts, USA
| | - Ryan Solinsky
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Laboratory, Spaulding Rehabilitation Hospital, Cambridge, Massachusetts, USA
| | - J. Andrew Taylor
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Laboratory, Spaulding Rehabilitation Hospital, Cambridge, Massachusetts, USA
| |
Collapse
|
57
|
Braunstein PW, Horovitz DJ, Hampton AM, Hollis F, Newman LA, Enos RT, McQuail JA. Daily fluctuations in blood glucose with normal aging are inversely related to hippocampal synaptic mitochondrial proteins. AGING BRAIN 2024; 5:100116. [PMID: 38596458 PMCID: PMC11002859 DOI: 10.1016/j.nbas.2024.100116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
Defective brain glucose utilization is a hallmark of Alzheimer's disease (AD) while Type II diabetes and elevated blood glucose escalate the risk for AD in later life. Isolating contributions of normal aging from coincident metabolic or brain diseases could lead to refined approaches to manage specific health risks and optimize treatments targeted to susceptible older individuals. We evaluated metabolic, neuroendocrine, and neurobiological differences between young adult (6 months) and aged (24 months) male rats. Compared to young adults, blood glucose was significantly greater in aged rats at the start of the dark phase of the day but not during the light phase. When challenged with physical restraint, a potent stressor, aged rats effected no change in blood glucose whereas blood glucose increased in young adults. Tissues were evaluated for markers of oxidative phosphorylation (OXPHOS), neuronal glucose transport, and synapses. Outright differences in protein levels between age groups were not evident, but circadian blood glucose was inversely related to OXPHOS proteins in hippocampal synaptosomes, independent of age. The neuronal glucose transporter, GLUT3, was positively associated with circadian blood glucose in young adults whereas aged rats tended to show the opposite trend. Our data demonstrate aging increases daily fluctuations in blood glucose and, at the level of individual differences, negatively associates with proteins related to synaptic OXPHOS. Our findings imply that glucose dyshomeostasis may exacerbate metabolic aspects of synaptic dysfunction that contribute to risk for age-related brain disorders.
Collapse
Affiliation(s)
- Paul W. Braunstein
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - David J. Horovitz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | | | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Lori A. Newman
- Department of Psychological Science, Vassar College, Poughkeepsie, NY, USA
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Joseph A. McQuail
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| |
Collapse
|
58
|
Asthana P, Wong HLX. Preventing obesity, insulin resistance and type 2 diabetes by targeting MT1-MMP. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167081. [PMID: 38367902 DOI: 10.1016/j.bbadis.2024.167081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Obesity is one of the predominant risk factors for type 2 diabetes. Despite all the modern advances in medicine, an effective drug treatment for obesity without overt side effects has not yet been found. The discovery of growth and differentiation factor 15 (GDF15), an appetite-regulating hormone, created hopes for the treatment of obesity. However, an insufficient understanding of the physiological regulation of GDF15 has been a major obstacle to mitigating GDF15-centric treatment of obesity. Our recent studies revealed how a series of proteolytic events predominantly mediated by membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP14), a key cell-surface metalloproteinase involved in extracellular remodeling, contribute to the pathogenesis of metabolic disorders, including obesity and diabetes. The MT1-MMP-mediated cleavage of the GDNF family receptor-α-like (GFRAL), a key neuronal receptor of GDF15, controls the satiety center in the hindbrain, thereby regulating non-homeostatic appetite and bodyweight changes. Furthermore, increased activation of MT1-MMP does not only lead to increased risk of obesity, but also causes age-associated insulin resistance by cleaving Insulin Receptor in major metabolic tissues. Importantly, inhibition of MT1-MMP effectively protects against obesity and diabetes, revealing the therapeutic potential of targeting MT1-MMP for the management of metabolic disorders.
Collapse
Affiliation(s)
- Pallavi Asthana
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | | |
Collapse
|
59
|
Yoshiko A, Shiozawa K, Niwa S, Takahashi H, Koike T, Watanabe K, Katayama K, Akima H. Association of skeletal muscle oxidative capacity with muscle function, sarcopenia-related exercise performance, and intramuscular adipose tissue in older adults. GeroScience 2024; 46:2715-2727. [PMID: 38153667 PMCID: PMC10828458 DOI: 10.1007/s11357-023-01043-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023] Open
Abstract
Muscle function and exercise performance measures, such as muscle endurance capacity, maximal strength, chair stand score, gait speed, and Timed Up and Go score, are evaluated to diagnose sarcopenia and frailty in older individuals. Furthermore, intramuscular adipose tissue (IntraMAT) content increases with age. Skeletal muscle oxidative capacity determines muscle metabolism and maintains muscle performance. This study aimed to investigate the association of skeletal muscle oxidative capacity with muscle function, exercise performance, and IntraMAT content in older individuals. Thirteen older men and women participated in this study. Skeletal muscle oxidative capacity was assessed by the recovery speed of muscle oxygen saturation after exercise using near-infrared spectroscopy from the medial gastrocnemius. We assessed two muscle functions, peak torque and time to task failure, and four sarcopenia-related exercise performances: handgrip strength, gait speed, 30-s chair stand, and Timed Up and Go. The IntraMAT content was measured using axial magnetic resonance imaging. The results showed a relationship between skeletal muscle oxidative capacity and gait speed but not with muscle functions and other exercise performance measures. Skeletal muscle oxidative capacity was not related to IntraMAT content. Skeletal muscle oxidative capacity, which may be indicative of the capacity of muscle energy production in the mitochondria, is related to locomotive functions but not to other functional parameters or skeletal fat infiltration.
Collapse
Affiliation(s)
- Akito Yoshiko
- Faculty of Liberal Arts and Sciences, Chukyo University, Toyota, Japan.
| | - Kana Shiozawa
- Department of Exercise and Sports Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Shiori Niwa
- Department of Nursing, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hideyuki Takahashi
- Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Teruhiko Koike
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kohei Watanabe
- School of Health and Sport Sciences, Chukyo University, Toyota, Japan
| | - Keisho Katayama
- Department of Exercise and Sports Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
| | - Hiroshi Akima
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
- Graduate School of Education and Human Development, Nagoya University, Nagoya, Japan
| |
Collapse
|
60
|
Miller MJ, Gries KJ, Marcotte GR, Ryan Z, Strub MD, Kunz HE, Arendt BK, Dasari S, Ebert SM, Adams CM, Lanza IR. Human myofiber-enriched aging-induced lncRNA FRAIL1 promotes loss of skeletal muscle function. Aging Cell 2024; 23:e14097. [PMID: 38297807 PMCID: PMC11019130 DOI: 10.1111/acel.14097] [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: 10/05/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024] Open
Abstract
The loss of skeletal muscle mass during aging is a significant health concern linked to adverse outcomes in older individuals. Understanding the molecular basis of age-related muscle loss is crucial for developing strategies to combat this debilitating condition. Long noncoding RNAs (lncRNAs) are a largely uncharacterized class of biomolecules that have been implicated in cellular homeostasis and dysfunction across a many tissues and cell types. To identify lncRNAs that might contribute to skeletal muscle aging, we screened for lncRNAs whose expression was altered in vastus lateralis muscle from older compared to young adults. We identified FRAIL1 as an aging-induced lncRNA with high abundance in human skeletal muscle. In healthy young and older adults, skeletal muscle FRAIL1 was increased with age in conjunction with lower muscle function. Forced expression of FRAIL1 in mouse tibialis anterior muscle elicits a dose-dependent reduction in skeletal muscle fiber size that is independent of changes in muscle fiber type. Furthermore, this reduction in muscle size is dependent on an intact region of FRAIL1 that is highly conserved across non-human primates. Unbiased transcriptional and proteomic profiling of the effects of FRAIL1 expression in mouse skeletal muscle revealed widespread changes in mRNA and protein abundance that recapitulate age-related changes in pathways and processes that are known to be altered in aging skeletal muscle. Taken together, these findings shed light on the intricate molecular mechanisms underlying skeletal muscle aging and implicate FRAIL1 in age-related skeletal muscle phenotypes.
Collapse
Affiliation(s)
- Matthew J. Miller
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- University of IowaIowa CityIowaUSA
| | | | - George R. Marcotte
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- University of IowaIowa CityIowaUSA
| | - Zachary Ryan
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | | | - Hawley E. Kunz
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | | | - Surendra Dasari
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Scott M. Ebert
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Emmyon, Inc.RochesterMinnesotaUSA
| | - Christopher M. Adams
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Emmyon, Inc.RochesterMinnesotaUSA
| | - Ian R. Lanza
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
61
|
Rathor L, Curry S, Park Y, McElroy T, Robles B, Sheng Y, Chen WW, Min K, Xiao R, Lee MH, Han SM. Mitochondrial stress in GABAergic neurons non-cell autonomously regulates organismal health and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585932. [PMID: 38585797 PMCID: PMC10996468 DOI: 10.1101/2024.03.20.585932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Mitochondrial stress within the nervous system can trigger non-cell autonomous responses in peripheral tissues. However, the specific neurons involved and their impact on organismal aging and health have remained incompletely understood. Here, we demonstrate that mitochondrial stress in γ-aminobutyric acid-producing (GABAergic) neurons in Caenorhabditis elegans ( C. elegans ) is sufficient to significantly alter organismal lifespan, stress tolerance, and reproductive capabilities. This mitochondrial stress also leads to significant changes in mitochondrial mass, energy production, and levels of reactive oxygen species (ROS). DAF-16/FoxO activity is enhanced by GABAergic neuronal mitochondrial stress and mediates the induction of these non-cell-autonomous effects. Moreover, our findings indicate that GABA signaling operates within the same pathway as mitochondrial stress in GABAergic neurons, resulting in non-cell-autonomous alterations in organismal stress tolerance and longevity. In summary, these data suggest the crucial role of GABAergic neurons in detecting mitochondrial stress and orchestrating non-cell-autonomous changes throughout the organism.
Collapse
|
62
|
Oba K, Ishikawa J, Tamura Y, Fujita Y, Ito M, Iizuka A, Fujiwara Y, Kodera R, Toyoshima K, Chiba Y, Tanaka M, Araki A. Serum Growth Differentiation Factor 15 Levels Predict the Incidence of Frailty among Patients with Cardiometabolic Diseases. Gerontology 2024; 70:517-525. [PMID: 38286122 DOI: 10.1159/000536150] [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: 05/04/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Frailty is a crucial health issue among older adults. Growth differentiation factor 15 (GDF15) is associated with inflammation, oxidative stress, insulin resistance, and mitochondrial dysfunction, which are possible pathogeneses of frailty. However, few longitudinal studies have investigated the association between GDF15 and the incidence of frailty. Therefore, we investigated whether high serum GDF15 levels are associated with the incidence of frailty. METHODS A total of 175 older adults (mean age: 77 ± 6 years; 63% women) with cardiometabolic diseases and no frailty out of the two criteria at baseline participated. Individuals with severe renal impairment or severe cognitive impairment were excluded. Serum GDF15 levels were measured at baseline. Patients were asked to assess frailty status at baseline and annually during follow-up using the modified version of the Cardiovascular Health Study (mCHS) and the Kihon Checklist (KCL). We examined the association between GDF15 tertiles and each frailty measure during follow-up (median 38-39 months). In the multivariate Cox regression analysis, with the GDF15 tertile groups as the explanatory variables, hazard ratios (HRs) and 95% confidence intervals (CIs) for incident frailty were calculated after adjusting for covariates and using the lowest tertile group as the reference. RESULTS During the follow-up period, 25.6% and 34.0% of patients developed frailty, as defined by the mCHS and KCL, respectively. The highest GDF15 tertile group had a significantly higher incidence of mCHS- or KCL-defined frailty than the lowest GDF15 tertile group. Multivariate Cox regression analysis revealed that the adjusted HRs for incident mCHS- and KCL-defined frailty in the highest GDF15 tertile group were 3.9 (95% CI: 1.3-12.0) and 2.7 (95% CI: 1.1-6.9), respectively. CONCLUSION High serum GDF15 levels predicted the incidence of frailty among older adults with cardiometabolic diseases and could be an effective marker of the risk for frailty in interventions aimed at preventing frailty, such as exercise and nutrition.
Collapse
Affiliation(s)
- Kazuhito Oba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Joji Ishikawa
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yasunori Fujita
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masafumi Ito
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Ai Iizuka
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Research Team for Social Participation and Community, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Remi Kodera
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kenji Toyoshima
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yuko Chiba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masashi Tanaka
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
63
|
Yang L, Zhang Z, Zhen Y, Feng J, Chen J, Song G. SIRT3 rs11246020 Polymorphism Associated Postprandial Triglyceride Dysmetabolism. Diabetes Metab Syndr Obes 2024; 17:1279-1288. [PMID: 38496003 PMCID: PMC10944304 DOI: 10.2147/dmso.s450962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Purpose Energy metabolism is regulated by SIRT3, no research has been done on the connection between lipid metabolism in the oral fat test and SIRT3 polymorphism. Thus, we conducted a case-control study to investigate the connection between postprandial lipid and SIRT3 polymorphism. Patients and Methods 402 non-obese Chinese subjects were enrolled and their postprandial lipid response to oral fat tolerance test (OFTT) was observed to understand the relationship between rs11246020 gene and postprandial triglyceride metabolism. Results In a binary logic regression model, a protective effect of the T allele of the rs11246020 SIRT3 for postprandial hypertriglyceridemia was shown (OR=0.417, 95% CI = 0.219-0.794, p=0.008). Compared to the CC genotype, individuals with the TT+CT variant of the rs11246020 SIRT3 gene demonstrated significantly lower levels of homeostasis model assessment of insulin resistance (HOMA-IR) (p=0.04), postprandial plasma glucose (PPG) (p=0.037), fasting plasma glucose (FPG) (p=0.02), and 4-hour triglyceridemia (Tg) (p=0.032). Conclusion The C allele of rs11246020 SIRT3 gene may be a risk factor to increased possibility of postprandial triglyceridemia after an oral fat test, which involved in the mechanism of glucose and insulin metabolism.
Collapse
Affiliation(s)
- Liqun Yang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, People’s Republic of China
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, Hebei Province, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Zhimei Zhang
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Yunfeng Zhen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Jing Feng
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Jinhu Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, People’s Republic of China
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, Hebei Province, People’s Republic of China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei Province, People’s Republic of China
| |
Collapse
|
64
|
Szablewski L. Insulin Resistance: The Increased Risk of Cancers. Curr Oncol 2024; 31:998-1027. [PMID: 38392069 PMCID: PMC10888119 DOI: 10.3390/curroncol31020075] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/15/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Insulin resistance, also known as impaired insulin sensitivity, is the result of a decreased reaction of insulin signaling to blood glucose levels. This state is observed when muscle cells, adipose tissue, and liver cells, improperly respond to a particular concentration of insulin. Insulin resistance and related increased plasma insulin levels (hyperinsulinemia) may cause metabolic impairments, which are pathological states observed in obesity and type 2 diabetes mellitus. Observations of cancer patients confirm that hyperinsulinemia is a major factor influencing obesity, type 2 diabetes, and cancer. Obesity and diabetes have been reported as risks of the initiation, progression, and metastasis of several cancers. However, both of the aforementioned pathologies may independently and additionally increase the cancer risk. The state of metabolic disorders observed in cancer patients is associated with poor outcomes of cancer treatment. For example, patients suffering from metabolic disorders have higher cancer recurrence rates and their overall survival is reduced. In these associations between insulin resistance and cancer risk, an overview of the various pathogenic mechanisms that play a role in the development of cancer is discussed.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego 5 Str., 02-004 Warsaw, Poland
| |
Collapse
|
65
|
Gonzalez-Armenta JL, Bergstrom J, Lee J, Furdui CM, Nicklas BJ, Molina AJA. Serum factors mediate changes in mitochondrial bioenergetics associated with diet and exercise interventions. GeroScience 2024; 46:349-365. [PMID: 37368157 PMCID: PMC10828137 DOI: 10.1007/s11357-023-00855-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: 04/27/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Mitochondrial improvements resulting from behavioral interventions, such as diet and exercise, are systemic and apparent across multiple tissues. Here, we test the hypothesis that factors present in serum, and therefore circulating throughout the body, can mediate changes in mitochondrial function in response to intervention. To investigate this, we used stored serum from a clinical trial comparing resistance training (RT) and RT plus caloric restriction (RT + CR) to examine effects of blood borne circulating factors on myoblasts in vitro. We report that exposure to dilute serum is sufficient to mediate bioenergetic benefits of these interventions. Additionally, serum-mediated bioenergetic changes can differentiate between interventions, recapitulate sex differences in bioenergetic responses, and is linked to improvements in physical function and inflammation. Using metabolomics, we identified circulating factors associated with changes in mitochondrial bioenergetics and the effects of interventions. This study provides new evidence that circulating factors play a role in the beneficial effects of interventions that improve healthspan among older adults. Understanding the factors that drive improvements in mitochondrial function is a key step towards predicting intervention outcomes and developing strategies to countermand systemic age-related bioenergetic decline.
Collapse
Affiliation(s)
- Jenny L Gonzalez-Armenta
- Section On Gerontology and Geriatrics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jaclyn Bergstrom
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, MC 0665, La Jolla, CA, 92093-0665, USA
| | - Jingyun Lee
- Proteomics and Metabolomics Shared Resource, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Section On Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barbara J Nicklas
- Section On Gerontology and Geriatrics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony J A Molina
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, MC 0665, La Jolla, CA, 92093-0665, USA.
| |
Collapse
|
66
|
Wang D, Zhang G, Yu Y, Zhang Z. Imaging of Sarcopenia in Type 2 Diabetes Mellitus. Clin Interv Aging 2024; 19:141-151. [PMID: 38292460 PMCID: PMC10826713 DOI: 10.2147/cia.s443572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Sarcopenia is an age-related condition characterized by the loss of skeletal muscle mass, muscular strength, and muscle function. In older adults, type 2 diabetes mellitus (T2DM) constitutes a significant health burden. Skeletal muscle damage and deterioration have emerged as novel chronic complications in patients with diabetes, often linked to their increased longevity. Diabetic sarcopenia has been associated with increased rates of hospitalization, cardiovascular events, and mortality. Nevertheless, effectively managing metabolic disorders in patients with T2DM through appropriate therapeutic interventions could potentially mitigate the risk of sarcopenia. Utilizing imaging technologies holds substantial clinical significance in the early detection of skeletal muscle mass alterations associated with sarcopenia. Such detection is pivotal for arresting disease progression and preserving patients' quality of life. These imaging modalities offer reproducible and consistent patterns over time, as they all provide varying degrees of quantitative data. This review primarily delves into the application of dual-energy X-ray absorptiometry, computed tomography, magnetic resonance imaging, and ultrasound for both qualitative and quantitative assessments of muscle mass in patients with T2DM. It also juxtaposes the merits and limitations of these four techniques. By understanding the nuances of each method, clinicians can discern how best to apply them in diverse clinical scenarios.
Collapse
Affiliation(s)
- Dingyue Wang
- Department of Ultrasound, the First Affiliated Hospital China Medical University, Shenyang City, Liaoning Province, 110001, People’s Republic of China
| | - Gaosen Zhang
- Department of Ultrasound, the First Affiliated Hospital China Medical University, Shenyang City, Liaoning Province, 110001, People’s Republic of China
| | - Yana Yu
- Department of Ultrasound, the First Affiliated Hospital China Medical University, Shenyang City, Liaoning Province, 110001, People’s Republic of China
| | - Zhen Zhang
- Department of Ultrasound, the First Affiliated Hospital China Medical University, Shenyang City, Liaoning Province, 110001, People’s Republic of China
| |
Collapse
|
67
|
Jha D, Prajapati SK, Deb PK, Jaiswal M, Mazumder PM. Madhuca longifolia-hydro-ethanolic-fraction reverses mitochondrial dysfunction and modulates selective GLUT expression in diabetic mice fed with high fat diet. Mol Biol Rep 2024; 51:209. [PMID: 38270737 DOI: 10.1007/s11033-023-08962-9] [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: 07/25/2023] [Accepted: 10/30/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Metabolic disorder is characterized as chronic low-grade inflammation which elevates the systemic inflammatory markers. The proposed hypothesis behind this includes occurrence of hypoxia due to intake of high fat diet leading to oxidative stress and mitochondrial dysfunction. AIM In the present work our aim was to elucidate the possible mechanism of action of hydroethanolic fraction of M. longifolia leaves against the metabolic disorder. METHOD AND RESULTS In the present investigation, effect of Madhuca longifolia hydroethanolic fraction (MLHEF) on HFD induced obesity and diabetes through mitochondrial action and selective GLUT expression has been studied. In present work, it was observed that HFD (50% of diet) on chronic administration aggravates the metabolic problems by causing reduced imbalanced oxidative stress, ATP production, and altered selective GLUT protein expression. Long term HFD administration reduced (p < 0.001) the SOD, CAT level significantly along with elevated liver function marker AST and ALT. MLHEF administration diminishes this oxidative stress. HFD administration also causes decreased ATP/ADP ratio owing to suppressed mitochondrial function and elevating LDH level. This oxidative imbalance further leads to dysregulated GLUT expression in hepatocytes, skeletal muscles and white adipose tissue. HFD leads to significant (p < 0.001) upregulation in GLUT 1 and 3 expression while significant (p < 0.001) downregulation in GLUT 2 and 4 expressions in WAT, liver and skeletal muscles. Administration of MLHEF significantly (p < 0.001) reduced the LDH level and also reduces the mitochondrial dysfunction. CONCLUSION Imbalances in GLUT levels were significantly reversed in order to maintain GLUT expression in tissues on the administration of MLHEF.
Collapse
Affiliation(s)
- Dhruv Jha
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India.
| | - Santosh Kumar Prajapati
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, 33613, USA
| | - Prashanta Kumar Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Mohit Jaiswal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| |
Collapse
|
68
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
69
|
Engin AB. Mechanism of Obesity-Related Lipotoxicity and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:131-166. [PMID: 39287851 DOI: 10.1007/978-3-031-63657-8_5] [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: 09/19/2024]
Abstract
The link between cellular exposure to fatty acid species and toxicity phenotypes remains poorly understood. However, structural characterization and functional profiling of human plasma free fatty acids (FFAs) analysis has revealed that FFAs are located either in the toxic cluster or in the cluster that is transcriptionally responsive to lipotoxic stress and creates genetic risk factors. Genome-wide short hairpin RNA screen has identified more than 350 genes modulating lipotoxicity. Hypertrophic adipocytes in obese adipose are both unable to expand further to store excess lipids in the diet and are resistant to the antilipolytic action of insulin. In addition to lipolysis, the inability of packaging the excess lipids into lipid droplets causes circulating fatty acids to reach toxic levels in non-adipose tissues. Deleterious effects of accumulated lipid in non-adipose tissues are known as lipotoxicity. Although triglycerides serve a storage function for long-chain non-esterified fatty acid and their products such as ceramide and diacylglycerols (DAGs), overloading of palmitic acid fraction of saturated fatty acids (SFAs) raises ceramide levels. The excess DAG and ceramide load create harmful effects on multiple organs and systems, inducing chronic inflammation in obesity. Thus, lipotoxic inflammation results in β cells death and pancreatic islets dysfunction. Endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and extracellular signal-regulated kinase (Erk) 1/2 signaling in adipocytes. However, palmitic acid-induced endoplasmic reticulum stress-c-Jun N-terminal kinase (JNK)-autophagy axis in hypertrophic adipocytes is a pro-survival mechanism against endoplasmic reticulum stress and cell death induced by SFAs. Endoplasmic reticulum-localized acyl-coenzyme A (CoA): glycerol-3-phosphate acyltransferase (GPAT) enzymes are mediators of lipotoxicity, and inhibiting these enzymes has therapeutic potential for lipotoxicity. Lipotoxicity increases the number of autophagosomes, which engulf palmitic acid, and thus suppress the autophagic turnover. Fatty acid desaturation promotes palmitate detoxification and storages into triglycerides. As therapeutic targets of glucolipotoxicity, in addition to caloric restriction and exercise, there are four different pharmacological approaches, which consist of metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, peroxisome proliferator-activated receptor-gamma (PPARγ) ligands thiazolidinediones, and chaperones are still used in clinical practice. Furthermore, induction of the brown fat-like phenotype with the mixture of eicosapentanoic acid and docosahexaenoic acid appears as a potential therapeutic application for treatment of lipotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
70
|
Yazıcı D, Demir SÇ, Sezer H. Insulin Resistance, Obesity, and Lipotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:391-430. [PMID: 39287860 DOI: 10.1007/978-3-031-63657-8_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Lipotoxicity, originally used to describe the destructive effects of excess fat accumulation on glucose metabolism, causes functional impairments in several metabolic pathways, both in adipose tissue and peripheral organs, like liver, heart, pancreas, and muscle. Ectopic lipid accumulation in the kidneys, liver, and heart has important clinical counterparts like diabetic nephropathy in type 2 diabetes mellitus, obesity-related glomerulopathy, nonalcoholic fatty liver disease, and cardiomyopathy. Insulin resistance due to lipotoxicity indirectly lead to reproductive system disorders, like polycystic ovary syndrome. Lipotoxicity has roles in insulin resistance and pancreatic beta-cell dysfunction. Increased circulating levels of lipids and the metabolic alterations in fatty acid utilization and intracellular signaling have been related to insulin resistance in muscle and liver. Different pathways, like novel protein kinase c pathways and the JNK-1 pathway, are involved as the mechanisms of how lipotoxicity leads to insulin resistance in nonadipose tissue organs, such as liver and muscle. Mitochondrial dysfunction plays a role in the pathogenesis of insulin resistance. Endoplasmic reticulum stress, through mainly increased oxidative stress, also plays an important role in the etiology of insulin resistance, especially seen in non-alcoholic fatty liver disease. Visceral adiposity and insulin resistance both increase the cardiometabolic risk, and lipotoxicity seems to play a crucial role in the pathophysiology of these associations.
Collapse
Affiliation(s)
- Dilek Yazıcı
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey.
| | - Selin Çakmak Demir
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| | - Havva Sezer
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| |
Collapse
|
71
|
Wang W, Xu K, Shang M, Li X, Tong X, Liu Z, Zhou L, Zheng S. The biological mechanism and emerging therapeutic interventions of liver aging. Int J Biol Sci 2024; 20:280-295. [PMID: 38164175 PMCID: PMC10750291 DOI: 10.7150/ijbs.87679] [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: 07/01/2023] [Accepted: 10/22/2023] [Indexed: 01/03/2024] Open
Abstract
Research on liver aging has become prominent and has attracted considerable interest in uncovering the mechanism and therapeutic targets of aging to expand lifespan. In addition, multi-omics studies are widely used to perform further mechanistic investigations on liver aging. In this review, we illustrate the changes that occur with aging in the liver, present the current models of liver aging, and emphasize existing multi-omics studies on liver aging. We integrated the multi-omics data of enrolled studies and reanalyzed them to identify key pathways and targets of liver aging. The results indicated that C-X-C motif chemokine ligand 9 (Cxcl9) was a regulator of liver aging. In addition, we provide a flowchart for liver aging research using multi-omics analysis and molecular experiments to help researchers conduct further research. Finally, we present emerging therapeutic treatments that prolong lifespan. In summary, using cells and animal models of liver aging, we can apply a multi-omics approach to find key metabolic pathways and target genes to mitigate the adverse effects of liver aging.
Collapse
Affiliation(s)
- Wenchao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Kangdi Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Mingge Shang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xiang Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xinyu Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310000, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310000, China
| |
Collapse
|
72
|
Bir A, Ghosh A, Müller WE, Ganguly A. Mitochondrial dysfunction and metabolic syndrome. METABOLIC SYNDROME 2024:157-172. [DOI: 10.1016/b978-0-323-85732-1.00043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
73
|
Duan J, Dong W, Wang G, Xiu W, Pu G, Xu J, Ye C, Zhang X, Zhu Y, Wang C. Senescence-associated 13-HODE production promotes age-related liver steatosis by directly inhibiting catalase activity. Nat Commun 2023; 14:8151. [PMID: 38071367 PMCID: PMC10710422 DOI: 10.1038/s41467-023-44026-z] [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: 04/01/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Aging is a major risk factor for metabolic disorders. Polyunsaturated fatty acid-derived bioactive lipids play critical roles as signaling molecules in metabolic processes. Nonetheless, their effects on age-related liver steatosis remain unknown. Here we show that senescent liver cells induce liver steatosis in a paracrine manner. Linoleic acid-derived 9-hydroxy-octadecadienoic acid (9-HODE) and 13-HODE increase in middle-aged (12-month-old) and aged (20-month-old) male mouse livers and conditioned medium from senescent hepatocytes and macrophages. Arachidonate 15-lipoxygenase, an enzyme for 13-HODE and 9-HODE production, is upregulated in senescent cells. A 9-HODE and 13-HODE mixture induces liver steatosis and activates SREBP1. Furthermore, catalase (CAT) is a direct target of 13-HODE, and its activity is decreased by 13-HODE. CAT overexpression reduces 13-HODE-induced liver steatosis and protects male mice against age-related liver steatosis. Therefore, 13-HODE produced by senescent hepatocytes and macrophages activates SREBP1 by directly inhibiting CAT activity and promotes liver steatosis.
Collapse
Affiliation(s)
- Jinjie Duan
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Wenhui Dong
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Guangyan Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Wenjing Xiu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Guangyin Pu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jingwen Xu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xu Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China.
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Chunjiong Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
74
|
Bartman CR, Faubert B, Rabinowitz JD, DeBerardinis RJ. Metabolic pathway analysis using stable isotopes in patients with cancer. Nat Rev Cancer 2023; 23:863-878. [PMID: 37907620 PMCID: PMC11161207 DOI: 10.1038/s41568-023-00632-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 11/02/2023]
Abstract
Metabolic reprogramming is central to malignant transformation and cancer cell growth. How tumours use nutrients and the relative rates of reprogrammed pathways are areas of intense investigation. Tumour metabolism is determined by a complex and incompletely defined combination of factors intrinsic and extrinsic to cancer cells. This complexity increases the value of assessing cancer metabolism in disease-relevant microenvironments, including in patients with cancer. Stable-isotope tracing is an informative, versatile method for probing tumour metabolism in vivo. It has been used extensively in preclinical models of cancer and, with increasing frequency, in patients with cancer. In this Review, we describe approaches for using in vivo isotope tracing to define fuel preferences and pathway engagement in tumours, along with some of the principles that have emerged from this work. Stable-isotope infusions reported so far have revealed that in humans, tumours use a diverse set of nutrients to supply central metabolic pathways, including the tricarboxylic acid cycle and amino acid synthesis. Emerging data suggest that some activities detected by stable-isotope tracing correlate with poor clinical outcomes and may drive cancer progression. We also discuss current challenges in isotope tracing, including comparisons of in vivo and in vitro models, and opportunities for future discovery in tumour metabolism.
Collapse
Affiliation(s)
- Caroline R Bartman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Brandon Faubert
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
75
|
Jhuo JY, Tong ZJ, Ku PH, Cheng HW, Wang HT. Acrolein induces mitochondrial dysfunction and insulin resistance in muscle and adipose tissues in vitro and in vivo. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122380. [PMID: 37625774 DOI: 10.1016/j.envpol.2023.122380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/05/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023]
Abstract
Type 2 diabetes mellitus (DM) is a common chronic condition characterized by persistent hyperglycemia and is associated with insulin resistance (IR) in critical glucose-consuming tissues, including skeletal muscle and adipose tissue. Oxidative stress and mitochondrial dysfunction are known to play key roles in IR. Acrolein is a reactive aldehyde found in the diet and environment that is generated as a fatty acid product through the glucose autooxidation process under hyperglycemic conditions. Our previous studies have shown that acrolein impairs insulin sensitivity in normal and diabetic mice, and this effect can be reversed by scavenging acrolein. This study demonstrated that acrolein increased oxidative stress and inhibited mitochondrial respiration in differentiated C2C12 myotubes and differentiated 3T3-L1 adipocytes. As a result, insulin signaling pathways were inhibited, leading to reduced glucose uptake. Treatment with acrolein scavengers, N-acetylcysteine, or carnosine ameliorated mitochondrial dysfunction and inhibited insulin signaling. Additionally, an increase in acrolein expression correlated with mitochondrial dysfunction in the muscle and adipose tissues of diabetic mice. These findings suggest that acrolein-induced mitochondrial dysfunction contributes to IR, and scavenging acrolein is a potential therapeutic approach for treating IR.
Collapse
Affiliation(s)
- Jia-Yu Jhuo
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Pei-Hsuan Ku
- Department of Life Sciences and the Institute of Genome Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
76
|
Garcia-Gaona E, García-Gregorio A, García-Jiménez C, López-Olaiz MA, Mendoza-Ramírez P, Fernandez-Guzman D, Pillado-Sánchez RA, Soto-Pacheco AD, Yareni-Zuñiga L, Sánchez-Parada MG, González-Santiago AE, Román-Pintos LM, Castañeda-Arellano R, Hernández-Ortega LD, Mercado-Sesma AR, Orozco-Luna FDJ, Villa-Angulo C, Villa-Angulo R, Baptista-Rosas RC. mtDNA Single-Nucleotide Variants Associated with Type 2 Diabetes. Curr Issues Mol Biol 2023; 45:8716-8732. [PMID: 37998725 PMCID: PMC10670651 DOI: 10.3390/cimb45110548] [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: 09/21/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic systemic disease with a complex etiology, characterized by insulin resistance and mitochondrial dysfunction in various cell tissues. To explore this relationship, we conducted a secondary analysis of complete mtDNA sequences from 1261 T2D patients and 1105 control individuals. Our findings revealed significant associations between certain single-nucleotide polymorphisms (SNPs) and T2D. Notably, the variants m.1438A>G (rs2001030) (controls: 32 [27.6%], T2D: 84 [72.4%]; OR: 2.46; 95%CI: 1.64-3.78; p < 0.001), m.14766C>T (rs193302980) (controls: 498 [36.9%], T2D: 853 [63.1%]; OR: 2.57, 95%CI: 2.18-3.04, p < 0.001), and m.16519T>C (rs3937033) (controls: 363 [43.4%], T2D: 474 [56.6%]; OR: 1.24, 95%CI: 1.05-1.47, p = 0.012) were significantly associated with the likelihood of developing diabetes. The variant m.16189T>C (rs28693675), which has been previously documented in several studies across diverse populations, showed no association with T2D in our analysis (controls: 148 [13.39] T2D: 171 [13.56%]; OR: 1.03; 95%CI: 0.815-1.31; p = 0.83). These results provide evidence suggesting a link between specific mtDNA polymorphisms and T2D, possibly related to association rules, topological patterns, and three-dimensional conformations associated with regions where changes occur, rather than specific point mutations in the sequence.
Collapse
Affiliation(s)
- Enrique Garcia-Gaona
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Puebla 72420, Mexico;
| | - Alhelí García-Gregorio
- Facultad de Enfermería Región Poza Rica-Tuxpan, Universidad Veracruzana, Veracruz 91700, Mexico;
| | - Camila García-Jiménez
- Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58000, Mexico;
| | | | - Paola Mendoza-Ramírez
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla 72420, Mexico;
| | | | | | - Axel David Soto-Pacheco
- Facultad de Medicina Extensión Los Mochis, Universidad Autónoma de Sinaloa, Sinaloa 81223, Mexico; (R.A.P.-S.); (A.D.S.-P.)
| | - Laura Yareni-Zuñiga
- Departamento de Ciencias de la Salud-Enfermedad como Proceso Individual, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (L.Y.-Z.); (L.M.R.-P.); (A.R.M.-S.)
| | - María Guadalupe Sánchez-Parada
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (M.G.S.-P.); (A.E.G.-S.); (R.C.-A.); (L.D.H.-O.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | - Ana Elizabeth González-Santiago
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (M.G.S.-P.); (A.E.G.-S.); (R.C.-A.); (L.D.H.-O.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | - Luis Miguel Román-Pintos
- Departamento de Ciencias de la Salud-Enfermedad como Proceso Individual, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (L.Y.-Z.); (L.M.R.-P.); (A.R.M.-S.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | - Rolando Castañeda-Arellano
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (M.G.S.-P.); (A.E.G.-S.); (R.C.-A.); (L.D.H.-O.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | - Luis Daniel Hernández-Ortega
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (M.G.S.-P.); (A.E.G.-S.); (R.C.-A.); (L.D.H.-O.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | - Arieh Roldán Mercado-Sesma
- Departamento de Ciencias de la Salud-Enfermedad como Proceso Individual, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (L.Y.-Z.); (L.M.R.-P.); (A.R.M.-S.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
| | | | - Carlos Villa-Angulo
- Laboratorio de Bioinformática y Biofotónica, Instituto de Ingeniería Universidad Autónoma de Baja California, Mexicali 21100, Mexico; (C.V.-A.); (R.V.-A.)
| | - Rafael Villa-Angulo
- Laboratorio de Bioinformática y Biofotónica, Instituto de Ingeniería Universidad Autónoma de Baja California, Mexicali 21100, Mexico; (C.V.-A.); (R.V.-A.)
| | - Raúl C. Baptista-Rosas
- Departamento de Ciencias de la Salud-Enfermedad como Proceso Individual, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá 45425, Mexico; (L.Y.-Z.); (L.M.R.-P.); (A.R.M.-S.)
- Centro de Investigación Multidisciplinaria en Salud, Universidad de Guadalajara, Tonalá 45425, Mexico
- Hospital General de Occidente, Secretaría de Salud Jalisco, Zapopan 45170, Mexico
| |
Collapse
|
77
|
Kasai S, Kokubu D, Mizukami H, Itoh K. Mitochondrial Reactive Oxygen Species, Insulin Resistance, and Nrf2-Mediated Oxidative Stress Response-Toward an Actionable Strategy for Anti-Aging. Biomolecules 2023; 13:1544. [PMID: 37892226 PMCID: PMC10605809 DOI: 10.3390/biom13101544] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Reactive oxygen species (ROS) are produced mainly by mitochondrial respiration and function as signaling molecules in the physiological range. However, ROS production is also associated with the pathogenesis of various diseases, including insulin resistance (IR) and type 2 diabetes (T2D). This review focuses on the etiology of IR and early events, especially mitochondrial ROS (mtROS) production in insulin-sensitive tissues. Importantly, IR and/or defective adipogenesis in the white adipose tissues (WAT) is thought to increase free fatty acid and ectopic lipid deposition to develop into systemic IR. Fatty acid and ceramide accumulation mediate coenzyme Q reduction and mtROS production in IR in the skeletal muscle, while coenzyme Q synthesis downregulation is also involved in mtROS production in the WAT. Obesity-related IR is associated with the downregulation of mitochondrial catabolism of branched-chain amino acids (BCAAs) in the WAT, and the accumulation of BCAA and its metabolites as biomarkers in the blood could reliably indicate future T2D. Transcription factor NF-E2-related factor 2 (Nrf2), which regulates antioxidant enzyme expression in response to oxidative stress, is downregulated in insulin-resistant tissues. However, Nrf2 inducers, such as sulforaphane, could restore Nrf2 and target gene expression and attenuate IR in multiple tissues, including the WAT.
Collapse
Affiliation(s)
- Shuya Kasai
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Daichi Kokubu
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Diet & Well-being Research Institute, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara 329-2762, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan;
| |
Collapse
|
78
|
Wang H, Akbari-Alavijeh S, Parhar RS, Gaugler R, Hashmi S. Partners in diabetes epidemic: A global perspective. World J Diabetes 2023; 14:1463-1477. [PMID: 37970124 PMCID: PMC10642420 DOI: 10.4239/wjd.v14.i10.1463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 09/01/2023] [Indexed: 10/09/2023] Open
Abstract
There is a recent increase in the worldwide prevalence of both obesity and diabetes. In this review we assessed insulin signaling, genetics, environment, lipid metabolism dysfunction and mitochondria as the major determinants in diabetes and to identify the potential mechanism of gut microbiota in diabetes diseases. We searched relevant articles, which have key information from laboratory experiments, epidemiological evidence, clinical trials, experimental models, meta-analysis and review articles, in PubMed, MEDLINE, EMBASE, Google scholars and Cochrane Controlled Trial Database. We selected 144 full-length articles that met our inclusion and exclusion criteria for complete assessment. We have briefly discussed these associations, challenges, and the need for further research to manage and treat diabetes more efficiently. Diabetes involves the complex network of physiological dysfunction that can be attributed to insulin signaling, genetics, environment, obesity, mitochondria and stress. In recent years, there are intriguing findings regarding gut microbiome as the important regulator of diabetes. Valid approaches are necessary for speeding medical advances but we should find a solution sooner given the burden of the metabolic disorder - What we need is a collaborative venture that may involve laboratories both in academia and industries for the scientific progress and its application for the diabetes control.
Collapse
Affiliation(s)
- Huan Wang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, Liaoning Province, China
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Safoura Akbari-Alavijeh
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Ranjit S Parhar
- Department of Biological and Medical Research, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Randy Gaugler
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Sarwar Hashmi
- Rutgers Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
- Research and Diagnostics, Ghazala and Sanya Hashmi Foundation, Holmdel, NJ 07733, United States
| |
Collapse
|
79
|
Cai F, Bezwada D, Cai L, Mahar R, Wu Z, Chang MC, Pachnis P, Yang C, Kelekar S, Gu W, Brooks B, Ko B, Vu HS, Mathews TP, Zacharias LG, Martin-Sandoval M, Do D, Oaxaca KC, Jin ES, Margulis V, Malloy CR, Merritt ME, DeBerardinis RJ. Comprehensive isotopomer analysis of glutamate and aspartate in small tissue samples. Cell Metab 2023; 35:1830-1843.e5. [PMID: 37611583 PMCID: PMC10732579 DOI: 10.1016/j.cmet.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 07/13/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
Stable isotopes are powerful tools to assess metabolism. 13C labeling is detected using nuclear magnetic resonance (NMR) spectroscopy or mass spectrometry (MS). MS has excellent sensitivity but generally cannot discriminate among different 13C positions (isotopomers), whereas NMR is less sensitive but reports some isotopomers. Here, we develop an MS method that reports all 16 aspartate and 32 glutamate isotopomers while requiring less than 1% of the sample used for NMR. This method discriminates between pathways that result in the same number of 13C labels in aspartate and glutamate, providing enhanced specificity over conventional MS. We demonstrate regional metabolic heterogeneity within human tumors, document the impact of fumarate hydratase (FH) deficiency in human renal cancers, and investigate the contributions of tricarboxylic acid (TCA) cycle turnover and CO2 recycling to isotope labeling in vivo. This method can accompany NMR or standard MS to provide outstanding sensitivity in isotope-labeling experiments, particularly in vivo.
Collapse
Affiliation(s)
- Feng Cai
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Divya Bezwada
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ling Cai
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Quantitative Biomedical Research Center, Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rohit Mahar
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Zheng Wu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mario C Chang
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Panayotis Pachnis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chendong Yang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sherwin Kelekar
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wen Gu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bailey Brooks
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bookyung Ko
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas P Mathews
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lauren G Zacharias
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Misty Martin-Sandoval
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Duyen Do
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - K Celeste Oaxaca
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Veterans Affairs North Texas Healthcare System, Dallas, TX 75216, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
80
|
Hyeon J, Lee J, Kim E, Lee HM, Kim KP, Shin J, Park HS, Lee YI, Nam CH. Vutiglabridin exerts anti-ageing effects in aged mice through alleviating age-related metabolic dysfunctions. Exp Gerontol 2023; 181:112269. [PMID: 37567452 DOI: 10.1016/j.exger.2023.112269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
BACKGROUND Ageing alters the ECM, leading to mitochondrial dysfunction and oxidative stress, which triggers an inflammatory response that exacerbates with age. Age-related changes impact satellite cells, affecting muscle regeneration, and the balance of proteins. Furthermore, ageing causes a decline in NAD+ levels, and alterations in fat metabolism that impact our health. These various metabolic issues become intricately intertwined with ageing, leading to a variety of individual-level diseases and profoundly affecting individuals' healthspan. Therefore, we hypothesize that vutiglabridin capable of alleviating these metabolic abnormalities will be able to ameliorate many of the problems associated with ageing. METHOD The efficacy of vutiglabridin, which alleviates metabolic issues by enhancing mitochondrial function, was assessed in aged mice treated with vutiglabridin and compared to untreated elderly mice. On young mice, vutiglabridin-treated aged mice, and non-treated aged mice, the Senescence-associated beta-galactosidase staining and q-PCR for ageing marker genes were carried out. Bulk RNA-seq was carried out on GA muscle, eWAT, and liver from each group of mice to compare differences in gene expression in various gene pathways. Blood from each group of mice was used to compare and analyze the ageing lipid profile. RESULTS SA-β-gal staining of eWAT, liver, kidney, and spleen of ageing mice showed that vutiglabridin had anti-ageing effects compared to the control group, and q-PCR of ageing marker genes including Cdkn1a and Cdkn2a in each tissue showed that vutiglabridin reduced the ageing process. In aged mice treated with vutiglabridin, GA muscle showed improved homeostasis compared to controls, eWAT showed restored insulin sensitivity and prevented FALC-induced inflammation, and liver showed reduced inflammation levels due to prevented TLO formation, improved mitochondrial complex I assembly, resulting in reduced ROS formation. Furthermore, blood lipid analysis revealed that ageing-related lipid profile was relieved in ageing mice treated with vutiglabridin versus the control group. CONCLUSION Vutiglabridin slows metabolic ageing mechanisms such as decreased insulin sensitivity, increased inflammation, and altered NAD+ metabolism in adipose tissue in mice experiments, while also retaining muscle homeostasis, which is deteriorated with age. It also improves the lipid profile in the blood and restores mitochondrial function in the liver to reduce ROS generation.
Collapse
Affiliation(s)
- Jooseung Hyeon
- Aging and Immunity Laboratory, Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Jihan Lee
- Aging and Immunity Laboratory, Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Eunju Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Republic of Korea; Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Hyeong Min Lee
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Republic of Korea; Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea; Glaceum Incorporation, Research Department, Suwon, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Republic of Korea; Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Jaejin Shin
- Glaceum Incorporation, Research Department, Suwon, Republic of Korea
| | - Hyung Soon Park
- Glaceum Incorporation, Research Department, Suwon, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, Department of Interdisciplinary Studies, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Chang-Hoon Nam
- Aging and Immunity Laboratory, Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea.
| |
Collapse
|
81
|
Hela F, Aguayo-Mazzucato C. Interaction between Autophagy and Senescence in Pancreatic Beta Cells. BIOLOGY 2023; 12:1205. [PMID: 37759604 PMCID: PMC10525299 DOI: 10.3390/biology12091205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023]
Abstract
Aging leads to an increase in cellular stress due to the fragility of the organism and the inability to cope with it. In this setting, there is a higher chance of developing different cardiometabolic diseases like diabetes. Cellular senescence and autophagy, both hallmarks of aging and stress-coping mechanisms, have gained increased attention for their role in the pathophysiology of diabetes. Studies show that impairing senescence dampens and even prevents diabetes while the role of autophagy is more contradictory, implying a context- and disease-stage-dependent effect. Reports show conflicting data about the effect of autophagy on senescence while the knowledge about this interaction in beta cells remains scarce. Elucidating this interaction between autophagy and senescence in pancreatic beta cells will lead to an identification of their respective roles and the extent of the effect each mechanism has on beta cells and open new horizons for developing novel therapeutic agents. To help illuminate this relationship we will review the latest findings of cellular senescence and autophagy with a special emphasis on pancreatic beta cells and diabetes.
Collapse
Affiliation(s)
| | - Cristina Aguayo-Mazzucato
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
82
|
Abounouh K, Tanouti IA, Ouladlahsen A, Tahiri M, Badre W, Dehbi H, Sarih M, Benjelloun S, Pineau P, Ezzikouri S. The peroxisome proliferator-activated receptor γ coactivator-1 alpha rs8192678 (Gly482Ser) variant and hepatitis B virus clearance. Infect Dis (Lond) 2023; 55:614-624. [PMID: 37376899 DOI: 10.1080/23744235.2023.2228403] [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: 01/09/2023] [Revised: 06/06/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Chronic hepatitis B virus (CHB) infection is still incurable a major public health problem. It is yet unclear how host genetic factors influence the development of HBV infection. The peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) has been shown to regulate hepatitis B virus (HBV). Several reports found that PPARGC1A variants are involved in a number of distinct liver diseases. Here we investigate whether the PPARGC1A rs8192678 (Gly482Ser) variant is involved in the spontaneous clearance of acute HBV infection and if it participates in chronic disease progression in Moroccan patients. METHODS Our study included 292 chronic hepatitis B (CHB) patients and 181 individuals who spontaneously cleared-HBV infection. We genotyped the rs8192678 SNP using a TaqMan allelic discrimination assay and then explored its association with spontaneous HBV clearance and CHB progression. RESULTS Our data showed that individuals carrying CT and TT genotypes were more likely to achieve spontaneous clearance (OR = 0.48, 95% CI (0.32-0.73), p = 0.00047; OR = 0.28, 95% CI (0.15-0.53), p = 0.00005, respectively). Subjects carrying the mutant allele T were more likely to achieve spontaneous clearance (OR = 0.51, 95% CI (0.38-0.67), P = 2.68E-06). However, when we investigated the impact of rs8192678 on the progression of liver diseases, we neither observe any influence (p > 0.05) nor found any significant association between ALT, AST, HBV viral loads, and the PPARGC1A rs8192678 genotypes in patients with CHB (p > 0.05). CONCLUSION Our result suggests that PPARGC1A rs8192678 may modulate acute HBV infection, and could therefore represent a potential predictive marker in the Moroccan population.
Collapse
Affiliation(s)
- Karima Abounouh
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
- Laboratory of Cellular and Molecular Pathology, Medical School, University Hassan II
| | - Ikram-Allah Tanouti
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Ahd Ouladlahsen
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Mohamed Tahiri
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Wafaa Badre
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Hind Dehbi
- Laboratory of Cellular and Molecular Pathology, Medical School, University Hassan II
| | - M'hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Soumaya Benjelloun
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Pascal Pineau
- Unité "Organisation Nucléaire et Oncogenèse", INSERM U993, Institut Pasteur, Paris, France
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
83
|
Dong H, Tsai SY. Mitochondrial Properties in Skeletal Muscle Fiber. Cells 2023; 12:2183. [PMID: 37681915 PMCID: PMC10486962 DOI: 10.3390/cells12172183] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Mitochondria are the primary source of energy production and are implicated in a wide range of biological processes in most eukaryotic cells. Skeletal muscle heavily relies on mitochondria for energy supplements. In addition to being a powerhouse, mitochondria evoke many functions in skeletal muscle, including regulating calcium and reactive oxygen species levels. A healthy mitochondria population is necessary for the preservation of skeletal muscle homeostasis, while mitochondria dysregulation is linked to numerous myopathies. In this review, we summarize the recent studies on mitochondria function and quality control in skeletal muscle, focusing mainly on in vivo studies of rodents and human subjects. With an emphasis on the interplay between mitochondrial functions concerning the muscle fiber type-specific phenotypes, we also discuss the effect of aging and exercise on the remodeling of skeletal muscle and mitochondria properties.
Collapse
Affiliation(s)
- Han Dong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
| | - Shih-Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
84
|
Wiseman RW, Brown CM, Beck TW, Brault JJ, Reinoso TR, Shi Y, Chase PB. Creatine Kinase Equilibration and ΔG ATP over an Extended Range of Physiological Conditions: Implications for Cellular Energetics, Signaling, and Muscle Performance. Int J Mol Sci 2023; 24:13244. [PMID: 37686064 PMCID: PMC10487889 DOI: 10.3390/ijms241713244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
In this report, we establish a straightforward method for estimating the equilibrium constant for the creatine kinase reaction (CK Keq″) over wide but physiologically and experimentally relevant ranges of pH, Mg2+ and temperature. Our empirical formula for CK Keq″ is based on experimental measurements. It can be used to estimate [ADP] when [ADP] is below the resolution of experimental measurements, a typical situation because [ADP] is on the order of micromolar concentrations in living cells and may be much lower in many in vitro experiments. Accurate prediction of [ADP] is essential for in vivo studies of cellular energetics and metabolism and for in vitro studies of ATP-dependent enzyme function under near-physiological conditions. With [ADP], we were able to obtain improved estimates of ΔGATP, necessitating the reinvestigation of previously reported ADP- and ΔGATP-dependent processes. Application to actomyosin force generation in muscle provides support for the hypothesis that, when [Pi] varies and pH is not altered, the maximum Ca2+-activated isometric force depends on ΔGATP in both living and permeabilized muscle preparations. Further analysis of the pH studies introduces a novel hypothesis around the role of submicromolar ADP in force generation.
Collapse
Affiliation(s)
- Robert Woodbury Wiseman
- Departments of Physiology and Radiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Caleb Micah Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Thomas Wesley Beck
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey John Brault
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Tyler Robert Reinoso
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yun Shi
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
85
|
Chen L, Zhou M, Li H, Liu D, Liao P, Zong Y, Zhang C, Zou W, Gao J. Mitochondrial heterogeneity in diseases. Signal Transduct Target Ther 2023; 8:311. [PMID: 37607925 PMCID: PMC10444818 DOI: 10.1038/s41392-023-01546-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 08/24/2023] Open
Abstract
As key organelles involved in cellular metabolism, mitochondria frequently undergo adaptive changes in morphology, components and functions in response to various environmental stresses and cellular demands. Previous studies of mitochondria research have gradually evolved, from focusing on morphological change analysis to systematic multiomics, thereby revealing the mitochondrial variation between cells or within the mitochondrial population within a single cell. The phenomenon of mitochondrial variation features is defined as mitochondrial heterogeneity. Moreover, mitochondrial heterogeneity has been reported to influence a variety of physiological processes, including tissue homeostasis, tissue repair, immunoregulation, and tumor progression. Here, we comprehensively review the mitochondrial heterogeneity in different tissues under pathological states, involving variant features of mitochondrial DNA, RNA, protein and lipid components. Then, the mechanisms that contribute to mitochondrial heterogeneity are also summarized, such as the mutation of the mitochondrial genome and the import of mitochondrial proteins that result in the heterogeneity of mitochondrial DNA and protein components. Additionally, multiple perspectives are investigated to better comprehend the mysteries of mitochondrial heterogeneity between cells. Finally, we summarize the prospective mitochondrial heterogeneity-targeting therapies in terms of alleviating mitochondrial oxidative damage, reducing mitochondrial carbon stress and enhancing mitochondrial biogenesis to relieve various pathological conditions. The possibility of recent technological advances in targeted mitochondrial gene editing is also discussed.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Zhou
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China.
| |
Collapse
|
86
|
Zeng X, Li L, Xia Z, Zou L, Kwok T, Su Y. Transcriptomic Analysis of Human Skeletal Muscle in Response to Aerobic Exercise and Protein Intake. Nutrients 2023; 15:3485. [PMID: 37571423 PMCID: PMC10421363 DOI: 10.3390/nu15153485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/29/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
This study aimed to provide a more comprehensive molecular insight into the effects of aerobic exercise (AE), protein intake (PI), and AE combined with PI on human skeletal muscle by comparing their transcriptomic profiles. Fourteen published datasets obtained from the Gene Expression Omnibus (GEO) database were used. The hub genes were identified in response to acute AE (ACTB, IL6), training AE (UBB, COL1A1), PI (EZH2), acute AE combined with PI (DDIT3), and training AE combined with PI (MYC). Both FOS and MYC were upregulated in response to acute AE, and they were, respectively, downregulated by higher PI and a combination of AE and PI. COL1A1 was upregulated by training AE but was downregulated by higher PI. Results from the gene set enrichment analysis (p < 0.05 and FDR < 25%) showed that AE and PI delivered their impacts on human skeletal muscle in analogous pathways, including aerobic respiration, mitochondrial complexes, extracellular matrix (ECM) remodeling, metabolic process, and immune/inflammatory responses, whereas, PI may attenuate the response of immune/inflammation and ECM remodeling which would be promoted by AE, irrespective of its types. Compared to PI alone, acute AE combined with PI would further promote protein turnover and synthesis, but suppress skeletal muscle contraction and movement.
Collapse
Affiliation(s)
- Xueqing Zeng
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China (Z.X.)
| | - Linghong Li
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China (Z.X.)
| | - Zhilin Xia
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China (Z.X.)
| | - Lianhong Zou
- Hunan Provincial Institute of Emergency Medicine, Hunan Provincial People’s Hospital, Changsha 410009, China
| | - Timothy Kwok
- Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi Su
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China (Z.X.)
| |
Collapse
|
87
|
Apostolova N, Vezza T, Muntane J, Rocha M, Víctor VM. Mitochondrial Dysfunction and Mitophagy in Type 2 Diabetes: Pathophysiology and Therapeutic Targets. Antioxid Redox Signal 2023; 39:278-320. [PMID: 36641637 DOI: 10.1089/ars.2022.0016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Significance: Type 2 diabetes mellitus, which is related to oxidative stress and mitochondrial dysfunction, is one of the most prevalent diseases in the world. In the past decade, alterations in autophagy have been shown to play a fundamental role in the development and control of type 2 diabetes. Further, mitophagy has been recognized as a key player in eliminating dysfunctional mitochondria in this disease. Recent Advances: Recently, much progress has been made in understanding the molecular events associated with oxidative stress, mitochondrial dysfunction, and alterations in autophagy and mitophagy in type 2 diabetes. Critical Issues: Despite increasing evidence of a relationship between mitochondrial dysfunction, oxidative stress, and alterations of autophagy and mitophagy and their role in the pathophysiolology of type 2 diabetes, effective therapeutic strategies to combat the disease through targeting mitochondria, autophagy, and mitophagy are yet to be implemented. Future Directions: This review provides a wide perspective of the existing literature concerning the complicated interplay between autophagy, mitophagy, and mitochondrial dysfunction in type 2 diabetes. Further, potential therapeutic targets based on these molecular mechanisms are explored. Antioxid. Redox Signal. 39, 278-320.
Collapse
Affiliation(s)
- Nadezda Apostolova
- Department of Pharmacology, University of Valencia, Valencia, Spain
- National Network of Biomedical Research on Hepatic and Digestive Diseases (CIBERehd), Valencia, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Teresa Vezza
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, FISABIO, Valencia, Spain
| | - Jordi Muntane
- National Network of Biomedical Research on Hepatic and Digestive Diseases (CIBERehd), Valencia, Spain
- Institute of Biomedicine of Seville (IBiS), Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| | - Milagros Rocha
- National Network of Biomedical Research on Hepatic and Digestive Diseases (CIBERehd), Valencia, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, FISABIO, Valencia, Spain
| | - Víctor M Víctor
- National Network of Biomedical Research on Hepatic and Digestive Diseases (CIBERehd), Valencia, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, FISABIO, Valencia, Spain
- Department of Physiology, University of Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
88
|
Zoico E, Saatchi T, Nori N, Mazzali G, Rizzatti V, Pizzi E, Fantin F, Giani A, Urbani S, Zamboni M. Senescent adipocytes as potential effectors of muscle cells dysfunction: An in vitro model. Exp Gerontol 2023; 179:112233. [PMID: 37321332 DOI: 10.1016/j.exger.2023.112233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 05/31/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Recently, there has been a growing body of evidence showing a negative effect of the white adipose tissue (WAT) dysfunction on the skeletal muscle function and quality. However, little is known about the effects of senescent adipocytes on muscle cells. Therefore, to explore potential mechanisms involved in age-related loss of muscle mass and function, we performed an in vitro experiment using conditioned medium obtained from cultures of mature and aged 3 T3-L1 adipocytes, as well as from cultures of dysfunctional adipocytes exposed to oxidative stress or high insulin doses, to treat C2C12 myocytes. The results from morphological measures indicated a significant decrease in diameter and fusion index of myotubes after treatment with medium of aged or stressed adipocytes. Aged and stressed adipocytes presented different morphological characteristics as well as a different gene expression profile of proinflammatory cytokines and ROS production. In myocytes treated with different adipocytes' conditioned media, we demonstrated a significant reduction of gene expression of myogenic differentiation markers as well as a significant increase of genes involved in atrophy. Finally, a significant reduction in protein synthesis as well as a significant increase of myostatin was found in muscle cells treated with medium of aged or stressed adipocytes compared to controls. In conclusion, these preliminary results suggest that aged adipocytes could influence negatively trophism, function and regenerative capacity of myocytes by a paracrine network of signaling.
Collapse
Affiliation(s)
- Elena Zoico
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Tanaz Saatchi
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy.
| | - Nicole Nori
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Gloria Mazzali
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Vanni Rizzatti
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Eleonora Pizzi
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| | - Francesco Fantin
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Anna Giani
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Silvia Urbani
- Department of Medicine, Geriatric Section, University of Verona, Verona, Italy
| | - Mauro Zamboni
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
89
|
Zhao F, Tomita M, Dutta A. Operational Modal Analysis of Near-Infrared Spectroscopy Measure of 2-Month Exercise Intervention Effects in Sedentary Older Adults with Diabetes and Cognitive Impairment. Brain Sci 2023; 13:1099. [PMID: 37509027 PMCID: PMC10377417 DOI: 10.3390/brainsci13071099] [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: 06/12/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The Global Burden of Disease Study (GBD 2019 Diseases and Injuries Collaborators) found that diabetes significantly increases the overall burden of disease, leading to a 24.4% increase in disability-adjusted life years. Persistently high glucose levels in diabetes can cause structural and functional changes in proteins throughout the body, and the accumulation of protein aggregates in the brain that can be associated with the progression of Alzheimer's Disease (AD). To address this burden in type 2 diabetes mellitus (T2DM), a combined aerobic and resistance exercise program was developed based on the recommendations of the American College of Sports Medicine. The prospectively registered clinical trials (NCT04626453, NCT04812288) involved two groups: an Intervention group of older sedentary adults with T2DM and a Control group of healthy older adults who could be either active or sedentary. The completion rate for the 2-month exercise program was high, with participants completing on an average of 89.14% of the exercise sessions. This indicated that the program was practical, feasible, and well tolerated, even during the COVID-19 pandemic. It was also safe, requiring minimal equipment and no supervision. Our paper presents portable near-infrared spectroscopy (NIRS) based measures that showed muscle oxygen saturation (SmO2), i.e., the balance between oxygen delivery and oxygen consumption in muscle, drop during bilateral heel rise task (BHR) and the 6 min walk task (6MWT) significantly (p < 0.05) changed at the post-intervention follow-up from the pre-intervention baseline in the T2DM Intervention group participants. Moreover, post-intervention changes from pre-intervention baseline for the prefrontal activation (both oxyhemoglobin and deoxyhemoglobin) showed statistically significant (p < 0.05, q < 0.05) effect at the right superior frontal gyrus, dorsolateral, during the Mini-Cog task. Here, operational modal analysis provided further insights into the 2-month exercise intervention effects on the very-low-frequency oscillations (<0.05 Hz) during the Mini-Cog task that improved post-intervention in the sedentary T2DM Intervention group from their pre-intervention baseline when compared to active healthy Control group. Then, the 6MWT distance significantly (p < 0.01) improved in the T2DM Intervention group at post-intervention follow-up from pre-intervention baseline that showed improved aerobic capacity and endurance. Our portable NIRS based measures have practical implications at the point of care for the therapists as they can monitor muscle and brain oxygenation changes during physical and cognitive tests to prescribe personalized physical exercise doses without triggering individual stress response, thereby, enhancing vascular health in T2DM.
Collapse
Affiliation(s)
- Fei Zhao
- Department of Rehabilitation Science, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Machiko Tomita
- Department of Rehabilitation Science, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Anirban Dutta
- School of Engineering, University of Lincoln, Lincoln LN67TS, UK
| |
Collapse
|
90
|
He LY, Li Y, Niu SQ, Bai J, Liu SJ, Guo JL. Polysaccharides from natural resource: ameliorate type 2 diabetes mellitus via regulation of oxidative stress network. Front Pharmacol 2023; 14:1184572. [PMID: 37497112 PMCID: PMC10367013 DOI: 10.3389/fphar.2023.1184572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/04/2023] [Indexed: 07/28/2023] Open
Abstract
Diabetes mellitus (DM) is a group of metabolic diseases characterized by hyperglycemia that can occur in children, adults, elderly people, and pregnant women. Oxidative stress is a significant adverse factor in the pathogenesis of DM, especially type 2 diabetes mellitus (T2DM), and metabolic syndrome. Natural polysaccharides are macromolecular compounds widely distributed in nature. Some polysaccharides derived from edible plants and microorganisms were reported as early as 10 years ago. However, the structural characterization of polysaccharides and their therapeutic mechanisms in diabetes are relatively shallow, limiting the application of polysaccharides. With further research, more natural polysaccharides have been reported to have antioxidant activity and therapeutic effects in diabetes, including plant polysaccharides, microbial polysaccharides, and polysaccharides from marine organisms and animals. Therefore, this paper summarizes the natural polysaccharides that have therapeutic potential for diabetes in the past 5 years, elucidating their pharmacological mechanisms and identified primary structures. It is expected to provide some reference for the application of polysaccharides, and provide a valuable resource for the development of new diabetic drugs.
Collapse
Affiliation(s)
- Li-Ying He
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shu-Qi Niu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing, China
| | - Jing Bai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si-Jing Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing, China
| | - Jin-Lin Guo
- Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing, China
| |
Collapse
|
91
|
Zapata Bustos R, Coletta DK, Galons JP, Davidson LB, Langlais PR, Funk JL, Willis WT, Mandarino LJ. Nonequilibrium thermodynamics and mitochondrial protein content predict insulin sensitivity and fuel selection during exercise in human skeletal muscle. Front Physiol 2023; 14:1208186. [PMID: 37485059 PMCID: PMC10361819 DOI: 10.3389/fphys.2023.1208186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction: Many investigators have attempted to define the molecular nature of changes responsible for insulin resistance in muscle, but a molecular approach may not consider the overall physiological context of muscle. Because the energetic state of ATP (ΔGATP) could affect the rate of insulin-stimulated, energy-consuming processes, the present study was undertaken to determine whether the thermodynamic state of skeletal muscle can partially explain insulin sensitivity and fuel selection independently of molecular changes. Methods: 31P-MRS was used with glucose clamps, exercise studies, muscle biopsies and proteomics to measure insulin sensitivity, thermodynamic variables, mitochondrial protein content, and aerobic capacity in 16 volunteers. Results: After showing calibrated 31P-MRS measurements conformed to a linear electrical circuit model of muscle nonequilibrium thermodynamics, we used these measurements in multiple stepwise regression against rates of insulin-stimulated glucose disposal and fuel oxidation. Multiple linear regression analyses showed 53% of the variance in insulin sensitivity was explained by 1) VO2max (p = 0.001) and the 2) slope of the relationship of ΔGATP with the rate of oxidative phosphorylation (p = 0.007). This slope represents conductance in the linear model (functional content of mitochondria). Mitochondrial protein content from proteomics was an independent predictor of fractional fat oxidation during mild exercise (R2 = 0.55, p = 0.001). Conclusion: Higher mitochondrial functional content is related to the ability of skeletal muscle to maintain a greater ΔGATP, which may lead to faster rates of insulin-stimulated processes. Mitochondrial protein content per se can explain fractional fat oxidation during mild exercise.
Collapse
Affiliation(s)
- Rocio Zapata Bustos
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
| | - Dawn K. Coletta
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
- Department of Physiology, The University of Arizona, Tucson, AZ, United States
| | - Jean-Philippe Galons
- Department of Medical Imaging, The University of Arizona, Tucson, AZ, United States
| | - Lisa B. Davidson
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
| | - Paul R. Langlais
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
| | - Janet L. Funk
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Wayne T. Willis
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
| | - Lawrence J. Mandarino
- Division of Endocrinology, Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
92
|
Maji RK, Czepukojc B, Scherer M, Tierling S, Cadenas C, Gianmoena K, Gasparoni N, Nordström K, Gasparoni G, Laggai S, Yang X, Sinha A, Ebert P, Falk-Paulsen M, Kinkley S, Hoppstädter J, Chung HR, Rosenstiel P, Hengstler JG, Walter J, Schulz MH, Kessler SM, Kiemer AK. Alterations in the hepatocyte epigenetic landscape in steatosis. Epigenetics Chromatin 2023; 16:30. [PMID: 37415213 DOI: 10.1186/s13072-023-00504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
Fatty liver disease or the accumulation of fat in the liver, has been reported to affect the global population. This comes with an increased risk for the development of fibrosis, cirrhosis, and hepatocellular carcinoma. Yet, little is known about the effects of a diet containing high fat and alcohol towards epigenetic aging, with respect to changes in transcriptional and epigenomic profiles. In this study, we took up a multi-omics approach and integrated gene expression, methylation signals, and chromatin signals to study the epigenomic effects of a high-fat and alcohol-containing diet on mouse hepatocytes. We identified four relevant gene network clusters that were associated with relevant pathways that promote steatosis. Using a machine learning approach, we predict specific transcription factors that might be responsible to modulate the functionally relevant clusters. Finally, we discover four additional CpG loci and validate aging-related differential CpG methylation. Differential CpG methylation linked to aging showed minimal overlap with altered methylation in steatosis.
Collapse
Affiliation(s)
- Ranjan Kumar Maji
- Institute for Cardiovascular Regeneration, Goethe-University, 60590, Frankfurt, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 60590, Frankfurt, Germany
| | - Beate Czepukojc
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Michael Scherer
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003, Barcelona, Spain
| | - Sascha Tierling
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Cristina Cadenas
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Kathrin Gianmoena
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Nina Gasparoni
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Karl Nordström
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Stephan Laggai
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Xinyi Yang
- Institute of Medical Bioinformatics and Biostatistics, Philipps University of Marburg, 35032, Marburg, Germany
| | - Anupam Sinha
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Peter Ebert
- Core Unit Bioinformatics, Medical Faculty, Heinrich Heine University, 40225, Düsseldorf, Germany
- Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, 66123, Saarbrücken, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Sarah Kinkley
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Ho-Ryun Chung
- Institute of Medical Bioinformatics and Biostatistics, Philipps University of Marburg, 35032, Marburg, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Jan G Hengstler
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe-University, 60590, Frankfurt, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 60590, Frankfurt, Germany.
- Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, 66123, Saarbrücken, Germany.
- Excellence Cluster on Multimodal Computing and Interaction, Saarland University, 66123, Saarbrücken, Germany.
| | - Sonja M Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany.
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Martin Luther University Halle-Wittenberg, Halle, Germany.
- Halle Research Centre for Drug Therapy (HRCDT), Halle, Germany.
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
93
|
Luo H, Zhu Z. Serum acylcarnitines levels as a potential predictor for gestational diabetes: a systematic review and meta-analysis. Front Public Health 2023; 11:1217237. [PMID: 37469690 PMCID: PMC10352491 DOI: 10.3389/fpubh.2023.1217237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Aims Gestational diabetes mellitus (GDM) stands as a prevalent obstetric complication bearing consequential health implications for both mother and child. While existing studies have probed the alterations in acylcarnitines during GDM, an updated systematic meta-analysis is needed to consolidate these findings. Hence, this study endeavours to furnish a comprehensive systematic review and meta-analysis delineating the association between acylcarnitines and GDM, aimed at bolstering diagnostic accuracy and preventive measures against GDM. Methods To extract pertinent studies for this meta-analysis, we meticulously scoured databases such as PubMed, Web of Science, Embase, and Cochrane Library up until May 2023. The inclusion criteria were studies contrasting plasma metabolomics between two cohorts: women diagnosed with GDM and normoglycemic pregnant women. Weighted mean differences (SMDs) and 95% confidence intervals (CIs) were calculated using random-effects models. The I2 index was employed to quantify heterogeneity amongst the studies. All meta-analyses were executed using Stata version 12.0. Results Our meta-analysis included eight studies encompassing 878 pregnant women. The analysis disclosed that relative to normoglycemic pregnant women, women with GDM exhibited significantly elevated levels of Short-Chain Acylcarnitines (SCAC) (SMD: 0.19, 95% CI: 0.02-0.36, I2 = 71.3%). No substantial difference was discerned in fasting total carnitine levels (SMD: 0.15, 95% CI: -0.16-0.31, I2 = 68.2%), medium-chain acylcarnitines (MCAC) (SMD: 0.08, 95% CI: -0.02-0.36, I2 = 79.0%), and long-chain acylcarnitines (LCAC) (SMD: 0.04, 95% CI: -0.06-0.15, I2 = 0%). Neither funnel plot assessment nor Egger's regression and Begg's rank correlation tests indicated any evidence of publication bias. Conclusion Our meta-analysis suggests that elevated levels of SCAC may heighten the risk of GDM onset. Given GDM's deleterious impact on pregnant women and their offspring, these findings underscore the clinical imperative of managing this condition. Early surveillance of plasma metabolomic profiles, particularly serum acylcarnitine concentrations, may equip clinicians with a valuable tool for timely diagnosis and intervention in GDM.
Collapse
Affiliation(s)
- Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zanlei Zhu
- Department of Respiratory and Critical Care Medicine, Ganzhou People’s Hospital (Ganzhou Hospital Affiliated to Nanchang University/Nanfang Medical University), Ganzhou, Jiangxi Province, China
| |
Collapse
|
94
|
Bahn YJ, Yadav H, Piaggi P, Abel BS, Gavrilova O, Springer DA, Papazoglou I, Zerfas PM, Skarulis MC, McPherron AC, Rane SG. CDK4-E2F3 signals enhance oxidative skeletal muscle fiber numbers and function to affect myogenesis and metabolism. J Clin Invest 2023; 133:e162479. [PMID: 37395281 PMCID: PMC10313363 DOI: 10.1172/jci162479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/19/2023] [Indexed: 07/04/2023] Open
Abstract
Understanding how skeletal muscle fiber proportions are regulated is vital to understanding muscle function. Oxidative and glycolytic skeletal muscle fibers differ in their contractile ability, mitochondrial activity, and metabolic properties. Fiber-type proportions vary in normal physiology and disease states, although the underlying mechanisms are unclear. In human skeletal muscle, we observed that markers of oxidative fibers and mitochondria correlated positively with expression levels of PPARGC1A and CDK4 and negatively with expression levels of CDKN2A, a locus significantly associated with type 2 diabetes. Mice expressing a constitutively active Cdk4 that cannot bind its inhibitor p16INK4a, a product of the CDKN2A locus, were protected from obesity and diabetes. Their muscles exhibited increased oxidative fibers, improved mitochondrial properties, and enhanced glucose uptake. In contrast, loss of Cdk4 or skeletal muscle-specific deletion of Cdk4's target, E2F3, depleted oxidative myofibers, deteriorated mitochondrial function, and reduced exercise capacity, while increasing diabetes susceptibility. E2F3 activated the mitochondrial sensor PPARGC1A in a Cdk4-dependent manner. CDK4, E2F3, and PPARGC1A levels correlated positively with exercise and fitness and negatively with adiposity, insulin resistance, and lipid accumulation in human and rodent muscle. All together, these findings provide mechanistic insight into regulation of skeletal muscle fiber-specification that is of relevance to metabolic and muscular diseases.
Collapse
Affiliation(s)
- Young Jae Bahn
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hariom Yadav
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona
| | - Brent S. Abel
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases
| | | | - Ioannis Papazoglou
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Monica C. Skarulis
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Alexandra C. McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Sushil G. Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
95
|
Abstract
Obesity, which has currently reached pandemic dimensions, is usually accompanied by diabetes mellitus type 2 (T2DM). These two conditions share common pathophysiological mechanisms. Adipose tissue secretes cytokines which are involved in inflammation and various endocrine functions. As for T2DM, it is characterized also by inflammation, mitochondrial dysfunction, and hyperinsulinemia. These conditions occur also in other diseases related to obesity and T2DM, like cardiovascular disease (CVD) and nonalcoholic fatty liver disease (NAFLD). Thus, management of obesity-related complications with lifestyle modification, anti-obesity drugs, and bariatric surgery, all contribute to improvement in any of these conditions. This review provides an overview of the literature addressing the association between obesity and T2DM, briefly discussing the pathophysiological mechanisms linking these conditions and outlining the management approach at the overlap of obesity and T2DM.
Collapse
Affiliation(s)
- Chrysoula Boutari
- Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Antea DeMarsilis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
96
|
Ispoglou T, Wilson O, McCullough D, Aldrich L, Ferentinos P, Lyall G, Stavropoulos-Kalinoglou A, Duckworth L, Brown MA, Sutton L, Potts AJ, Archbold V, Hargreaves J, McKenna J. A Narrative Review of Non-Pharmacological Strategies for Managing Sarcopenia in Older Adults with Cardiovascular and Metabolic Diseases. BIOLOGY 2023; 12:892. [PMID: 37508325 PMCID: PMC10376679 DOI: 10.3390/biology12070892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023]
Abstract
This narrative review examines the mechanisms underlying the development of cardiovascular disease (CVD) and metabolic diseases (MDs), along with their association with sarcopenia. Furthermore, non-pharmacological interventions to address sarcopenia in patients with these conditions are suggested. The significance of combined training in managing metabolic disease and secondary sarcopenia in type II diabetes mellitus is emphasized. Additionally, the potential benefits of resistance and aerobic training are explored. This review emphasises the role of nutrition in addressing sarcopenia in patients with CVD or MDs, focusing on strategies such as optimising protein intake, promoting plant-based protein sources, incorporating antioxidant-rich foods and omega-3 fatty acids and ensuring sufficient vitamin D levels. Moreover, the potential benefits of targeting gut microbiota through probiotics and prebiotic fibres in sarcopenic individuals are considered. Multidisciplinary approaches that integrate behavioural science are explored to enhance the uptake and sustainability of behaviour-based sarcopenia interventions. Future research should prioritise high-quality randomized controlled trials to refine exercise and nutritional interventions and investigate the incorporation of behavioural science into routine practices. Ultimately, a comprehensive and multifaceted approach is essential to improve health outcomes, well-being and quality of life in older adults with sarcopenia and coexisting cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
| | - Oliver Wilson
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Luke Aldrich
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Gemma Lyall
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | | | - Lauren Duckworth
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Meghan A Brown
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Louise Sutton
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Alexandra J Potts
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Victoria Archbold
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Jackie Hargreaves
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| | - Jim McKenna
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QQ, UK
| |
Collapse
|
97
|
Chen L, Qin G, Liu Y, Li M, Li Y, Guo LZ, Du L, Zheng W, Wu PC, Chuang YH, Wang X, Wang TD, Ho JAA, Liu TM. Label-free optical metabolic imaging of adipose tissues for prediabetes diagnosis. Theranostics 2023; 13:3550-3567. [PMID: 37441598 PMCID: PMC10334843 DOI: 10.7150/thno.82697] [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: 01/16/2023] [Accepted: 05/11/2023] [Indexed: 07/15/2023] Open
Abstract
Rationale: Prediabetes can be reversed through lifestyle intervention, but its main pathologic hallmark, insulin resistance (IR), cannot be detected as conveniently as blood glucose testing. In consequence, the diagnosis of prediabetes is often delayed until patients have hyperglycemia. Therefore, developing a less invasive diagnostic method for rapid IR evaluation will contribute to the prognosis of prediabetes. Adipose tissue is an endocrine organ that plays a crucial role in the development and progression of prediabetes. Label-free visualizing the prediabetic microenvironment of adipose tissues provides a less invasive alternative for the characterization of IR and inflammatory pathology. Methods: Here, we successfully identified the differentiable features of prediabetic adipose tissues by employing the metabolic imaging of three endogenous fluorophores NAD(P)H, FAD, and lipofuscin-like pigments. Results: We discovered that 1040-nm excited lipofuscin-like autofluorescence could mark the location of macrophages. This unique feature helps separate the metabolic fluorescence signals of macrophages from those of adipocytes. In prediabetes fat tissues with IR, we found only adipocytes exhibited a low redox ratio of metabolic fluorescence and high free NAD(P)H fraction a1. This differential signature disappears for mice who quit the high-fat diet or high-fat-high-sucrose diet and recover from IR. When mice have diabetic hyperglycemia and inflamed fat tissues, both adipocytes and macrophages possess this kind of metabolic change. As confirmed with RNA-seq analysis and histopathology evidence, the change in adipocyte's metabolic fluorescence could be an indicator or risk factor of prediabetic IR. Conclusion: Our study provides an innovative approach to diagnosing prediabetes, which sheds light on the strategy for diabetes prevention.
Collapse
Affiliation(s)
- Liping Chen
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Guihui Qin
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Moxin Li
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Yue Li
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Lun-Zhang Guo
- Institute of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Lidong Du
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Weiming Zheng
- Translational Medicine R&D Center, Zhuhai UM Science and Technology Research Institute, Zhuhai, China
| | - Pei-Chun Wu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- Department of Biochemical Science & Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Yueh-Hsun Chuang
- Department of Anesthesiology, National Taiwan University Hospital and College of Medicine, Taipei 10002, Taiwan
| | - Xiaoyan Wang
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| | - Tzung-Dau Wang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei 10002, Taiwan
| | - Ja-An Annie Ho
- Department of Biochemical Science & Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, China
| |
Collapse
|
98
|
Al-Sofiani ME, Asiri A, Alajmi S, Alkeridy W. Perspectives on Prediabetes and Aging. Endocrinol Metab Clin North Am 2023; 52:377-388. [PMID: 36948785 DOI: 10.1016/j.ecl.2022.10.011] [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] [Indexed: 02/18/2023]
Abstract
Diabetes prevention programs (DPPs) have been shown to effectively delay, and sometimes prevent, the progression from prediabetes to diabetes; however, labeling someone with prediabetes comes with potential negative psychological, financial, and self-perception consequences. Many older adults with prediabetes nowadays have a relatively "low-risk" form of prediabetes that rarely progresses to diabetes and may regress to normoglycemia. In this article, we review the impact of aging on glucose metabolism and provide a holistic approach to cases of prediabetes in older adults that maximizes the benefit-risk balance of interventions aimed at addressing prediabetes.
Collapse
Affiliation(s)
- Mohammed E Al-Sofiani
- Division of Endocrinology, Department of Internal Medicine, College of Medicine, King Saud University, Riyadh, Central Region, 12372, Saudi Arabia; Division of Endocrinology, Diabetes & Metabolism, The Johns Hopkins University, 1830 East Monument Street, Baltimore, MD 21287, USA.
| | - Alanood Asiri
- Division of Endocrinology, Department of Internal Medicine, College of Medicine, King Saud University, Riyadh, Central Region, 12372, Saudi Arabia
| | - Sarah Alajmi
- Division of Endocrinology, Department of Internal Medicine, College of Medicine, King Saud University, Riyadh, Central Region, 12372, Saudi Arabia
| | - Walid Alkeridy
- Department of Medicine, King Saud University, College of Medicine, Riyadh, Central Region, 12372, Saudi Arabia
| |
Collapse
|
99
|
Moellering DR, Smith-Johnston K, Kelley C, Sammy MJ, Benedict J, Brock G, Johnson J, Baskin KK, Jarjour WN, Belury MA, Reiser PJ, Nagareddy PR, Hanaoka BY. Association between skeletal muscle mitochondrial dysfunction and insulin resistance in patients with rheumatoid arthritis: a case-control study. Arthritis Res Ther 2023; 25:85. [PMID: 37210569 PMCID: PMC10199606 DOI: 10.1186/s13075-023-03065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Insulin resistance affects a substantial proportion of patients with rheumatoid arthritis (RA). Skeletal muscle mitochondrial dysfunction results in the accumulation of lipid intermediates that interfere with insulin signaling. We therefore sought to determine if lower oxidative phosphorylation and muscle mitochondrial content are associated with insulin resistance in patients with RA. METHODS This was a cross-sectional prospective study of RA patients. Matsuda index from the glucose tolerance test was used to estimate insulin sensitivity. Mitochondrial content was measured by citrate synthase (CS) activity in snap-frozen muscle samples. Mitochondrial function was measured by using high-resolution respirometry of permeabilized muscle fibers and electron transport chain complex IV enzyme kinetics in isolated mitochondrial subpopulations. RESULTS RA participants demonstrated lower insulin sensitivity as measured by the Matsuda index compared to controls [median 3.95 IQR (2.33, 5.64) vs. 7.17 (5.83, 7.75), p = 0.02]. There was lower muscle mitochondrial content among RA vs. controls [median 60 mU/mg IQR (45, 80) vs. 79 mU/mg (65, 97), p = 0.03]. Notably, OxPhos normalized to mitochondrial content was higher among RA vs. controls [mean difference (95% CI) = 0.14 (0.02, 0.26), p = 0.03], indicating a possible compensatory mechanism for lower mitochondrial content or lipid overload. Among RA participants, the activity of muscle CS activity was not correlated with the Matsuda index (ρ = - 0.05, p = 0.84), but it was positively correlated with self-reported (IPAQ) total MET-minutes/week (ρ = 0.44, p = 0.03) and Actigraph-measured time on physical activity (MET rate) (ρ = 0.47, p = 0.03). CONCLUSIONS Mitochondrial content and function were not associated with insulin sensitivity among participants with RA. However, our study demonstrates a significant association between muscle mitochondrial content and physical activity level, highlighting the potential for future exercise interventions that enhance mitochondrial efficiency in RA patients.
Collapse
Affiliation(s)
- Douglas R Moellering
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kelley Smith-Johnston
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christian Kelley
- Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melissa J Sammy
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Benedict
- Department of Biomedical Bioinformatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Guy Brock
- Department of Biomedical Bioinformatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jillian Johnson
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Kedryn K Baskin
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Wael N Jarjour
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Martha A Belury
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Prabhakara R Nagareddy
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Beatriz Y Hanaoka
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
100
|
Turkel I, Ozerklig B, Yılmaz M, Ulger O, Kubat GB, Tuncer M. Mitochondrial transplantation as a possible therapeutic option for sarcopenia. J Mol Med (Berl) 2023:10.1007/s00109-023-02326-3. [PMID: 37209146 DOI: 10.1007/s00109-023-02326-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/09/2023] [Accepted: 04/20/2023] [Indexed: 05/22/2023]
Abstract
With advancing age, the skeletal muscle phenotype is characterized by a progressive loss of mass, strength, and quality. This phenomenon, known as sarcopenia, has a negative impact on quality of life and increases the risk of morbidity and mortality in older adults. Accumulating evidence suggests that damaged and dysfunctional mitochondria play a critical role in the pathogenesis of sarcopenia. Lifestyle modifications, such as physical activity, exercise, and nutrition, as well as medical interventions with therapeutic agents, are effective in the management of sarcopenia and offer solutions to maintain and improve skeletal muscle health. Although a great deal of effort has been devoted to the identification of the best treatment option, these strategies are not sufficient to overcome sarcopenia. Recently, it has been reported that mitochondrial transplantation may be a possible therapeutic approach for the treatment of mitochondria-related pathological conditions such as ischemia, liver toxicity, kidney injury, cancer, and non-alcoholic fatty liver disease. Given the role of mitochondria in the function and metabolism of skeletal muscle, mitochondrial transplantation may be a possible option for the treatment of sarcopenia. In this review, we summarize the definition and characteristics of sarcopenia and molecular mechanisms associated with mitochondria that are known to contribute to sarcopenia. We also discuss mitochondrial transplantation as a possible option. Despite the progress made in the field of mitochondrial transplantation, further studies are needed to elucidate the role of mitochondrial transplantation in sarcopenia. KEY MESSAGES: Sarcopenia is the progressive loss of skeletal muscle mass, strength, and quality. Although the specific mechanisms that lead to sarcopenia are not fully understood, mitochondria have been identified as a key factor in the development of sarcopenia. Damaged and dysfunctional mitochondria initiate various cellular mediators and signaling pathways, which largely contribute to the age-related loss of skeletal muscle mass and strength. Mitochondrial transplantation has been reported to be a possible option for the treatment/prevention of several diseases. Mitochondrial transplantation may be a possible therapeutic option for improving skeletal muscle health and treating sarcopenia. Mitochondrial transplantation as a possible treatment option for sarcopenia.
Collapse
Affiliation(s)
- Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Merve Yılmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Health Sciences Institute, Health Sciences University, Ankara, Turkey
| | - Gokhan Burcin Kubat
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey.
- Department of Mitochondria and Cellular Research, Health Sciences Institute, Health Sciences University, Ankara, Turkey.
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|