1
|
Rogers K, Kiernan E, Partie M, Kenkel W. Impact of Cesarean Delivery on Reward Behavior and Neurodevelopment in Adult Prairie Vole Offspring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646614. [PMID: 40236166 PMCID: PMC11996564 DOI: 10.1101/2025.04.01.646614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Accumulating clinical evidence has shown that birth by Cesarean section (CS) is associated with a higher incidence of disorders involving the dysregulation of dopamine (DA), such as attention deficit-hyperactivity disorder, autism spectrum disorder, and obesity, compared to vaginal delivery (VD). The mesolimbic (ML) system encompasses DAergic neurons that modulate reward processes underlying learning, motivation, and food intake. Previous research has shown that there are lower levels of DA in the prefrontal cortex and higher in the nucleus accumbens (NAc) of CS offspring. Alterations in the ML-DA system as a consequence of birth via CS may impact behavioral response to rewarding stimuli, such as food. Thus, we aimed to ascertain the behavioral and neurodevelopmental outcomes relevant to food reward in CS prairie vole offspring. This study utilized conditioned place preference (CPP) testing to assess learning using context-dependent conditioning, operant conditioning to assess acquisition of a conditioned response and motivation to receive a reinforcer, and immunohistochemistry (IHC) to stain for tyrosine hydroxylase (TH) in the NAc. Behavioral results showed no difference in preference for the conditioned chamber during CPP testing between CS offspring and their VD counterparts. CS prairie vole offspring had a lower average break point during progressive-ratio testing compared to VD offspring, but no difference in response during fixed-ratio 1 or 3 testing. IHC results showed CS offspring had lower levels of TH-immunoreactivity in the NAc core and shell. These findings further support that delivery by CS has long-term neurodevelopmental effects, specifically in the brain's reward system, and that CS offspring have decreased motivation toward food reward independent of deficits in learning.
Collapse
|
2
|
Blok NB, Myronovych A, McMahon G, Bozadjieva-Kramer N, Seeley RJ. The evolution of steatosis and fibrosis in mice on a MASH-inducing diet and the effects of housing temperature. Am J Physiol Endocrinol Metab 2025; 328:E513-E523. [PMID: 39998384 DOI: 10.1152/ajpendo.00401.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/05/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Obesity induction in mice requires high-fat diet exposure. Although hepatic steatosis develops, progression to inflammation and fibrosis, as in humans, requires prolonged exposure and additional dietary factors. Immunosuppression at room temperature may slow this progression. We evaluated thermoneutrality's effect on metabolic dysfunction-associated steatohepatitis (MASH) development using a fibrosis-inducing MASH [Gubra-Amylin NASH (GAN)] diet. Mice were fed either a MASH or chow diet and housed at room temperature or thermoneutrality. MASH diet groups were euthanized monthly from 4 to 7 mo. Serum markers of hepatic function were analyzed, and liver histology assessed steatosis, inflammation, ballooning [nonalcoholic fatty liver disease activity score (NAS) score], and fibrosis via Picrosirius Red staining. MASH diet increased body weight, liver-to-body mass ratio, and hepatic fat, with no difference between housing conditions. Housing temperature had minimal effects on MASH. Serum markers and hepatic fibrosis were similar across groups. NAS score was lower at 4 mo in thermoneutral MASH mice but not by 7 mo. Thermoneutrality did not significantly impact MASH development. These findings, alongside existing literature, suggest thermoneutral housing does not consistently enhance MASH progression in GAN MASH-fed mice.NEW & NOTEWORTHY The development of MASH in mice-does housing temperature make a real difference?
Collapse
Affiliation(s)
- Neil B Blok
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Andriy Myronovych
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Garrett McMahon
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
3
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [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: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
4
|
Raun SH, Braun JL, Karavaeva I, Henriquez-Olguín C, Ali MS, Møller LLV, Gerhart-Hines Z, Fajardo VA, Richter EA, Sylow L. Mild Cold Stress at Ambient Temperature Elevates Muscle Calcium Cycling and Exercise Adaptations in Obese Female Mice. Endocrinology 2024; 165:bqae102. [PMID: 39136248 DOI: 10.1210/endocr/bqae102] [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: 04/09/2024] [Indexed: 08/28/2024]
Abstract
CONTEXT Housing temperature is a critical regulator of mouse metabolism and thermoneutral housing can improve model translation to humans. However, the impact of housing temperature on the ability of wheel running exercise training to rescue the detrimental effect of diet-induced obese mice is currently not fully understood. OBJECTIVE To investigate how housing temperature affects muscle metabolism in obese mice with regard to calcium handling and exercise training (ET) adaptations in skeletal muscle, and benefits of ET on adiposity and glucometabolic parameters. METHODS Lean or obese female mice were housed at standard ambient temperature (22 °C) or thermoneutrality (30 °C) with/without access to running wheels. The metabolic phenotype was investigated using glucose tolerance tests, indirect calorimetry, and body composition. Molecular muscle adaptations were measured using immunoblotting, qPCR, and spectrophotometric/fluorescent assays. RESULTS Obese female mice housed at 22 °C showed lower adiposity, lower circulating insulin levels, improved glucose tolerance, and elevated basal metabolic rate compared to 30 °C housing. Mice exposed to voluntary wheel running exhibited a larger fat loss and higher metabolic rate at 22 °C housing compared to thermoneutrality. In obese female mice, glucose tolerance improved after ET independent of housing temperature. Independent of diet and training, 22 °C housing increased skeletal muscle sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) activity. Additionally, housing at 22 °C elevated the induction of training-responsive muscle proteins in obese mice. CONCLUSION Our findings highlight that housing temperature significantly influences adiposity, insulin sensitivity, muscle physiology, and exercise adaptations in diet-induced obese female mice.
Collapse
Affiliation(s)
- Steffen H Raun
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jessica L Braun
- Muscle Plasticity in Health and Disease, Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, L2A 3A1, Canada
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Carlos Henriquez-Olguín
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 7501015, Chile
| | - Mona S Ali
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lisbeth L V Møller
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Val A Fajardo
- Muscle Plasticity in Health and Disease, Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, L2A 3A1, Canada
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen 2100, Denmark
| | - Lykke Sylow
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
5
|
Stojchevski R, Chandrasekaran P, Hadzi-Petrushev N, Mladenov M, Avtanski D. Adipose Tissue Dysfunction Related to Climate Change and Air Pollution: Understanding the Metabolic Consequences. Int J Mol Sci 2024; 25:7849. [PMID: 39063092 PMCID: PMC11277516 DOI: 10.3390/ijms25147849] [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: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity, a global pandemic, poses a major threat to healthcare systems worldwide. Adipose tissue, the energy-storing organ during excessive energy intake, functions as a thermoregulator, interacting with other tissues to regulate systemic metabolism. Specifically, brown adipose tissue (BAT) is positively associated with an increased resistance to obesity, due to its thermogenic function in the presence of uncoupled protein 1 (UCP1). Recently, studies on climate change and the influence of environmental pollutants on energy homeostasis and obesity have drawn increasing attention. The reciprocal relationship between increasing adiposity and increasing temperatures results in reduced adaptive thermogenesis, decreased physical activity, and increased carbon footprint production. In addition, the impact of climate change makes obese individuals more prone to developing type 2 diabetes mellitus (T2DM). An impaired response to heat stress, compromised vasodilation, and sweating increase the risk of diabetes-related comorbidities. This comprehensive review provides information about the effects of climate change on obesity and adipose tissue, the risk of T2DM development, and insights into the environmental pollutants causing adipose tissue dysfunction and obesity. The effects of altered dietary patterns on adiposity and adaptation strategies to mitigate the detrimental effects of climate change are also discussed.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10003, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10003, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
6
|
Kenkel WM, Ahmed S, Partie M, Rogers K. Delivery by cesarean section leads to heavier adult bodyweight in prairie voles (Microtus ochrogaster). Horm Behav 2024; 160:105499. [PMID: 38350334 PMCID: PMC10961198 DOI: 10.1016/j.yhbeh.2024.105499] [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: 09/12/2023] [Revised: 11/20/2023] [Accepted: 01/29/2024] [Indexed: 02/15/2024]
Abstract
Delivery by cesarean section now makes up 32.1 % of all births in the United States. Meta-analyses have estimated that delivery by cesarean section is associated with a > 50 % increased risk for childhood obesity by 5 years of age. While this association is independent of maternal obesity, breastfeeding, and heritable factors, studies in humans have been unable to test for a causal role of cesarean delivery in this regard. Here, we set out to use an animal model to experimentally test whether delivery by cesarean section would increase offspring weight in adulthood. Delivery by cesarean section may exert neurodevelopmental consequences by impacting hormones that are important at birth as well as during metabolic regulation in later life, such as oxytocin and vasopressin. The prairie vole (Microtus ochrogaster) has long been studied to investigate the roles of oxytocin and vasopressin in brain development and social behavior. Here, we establish that prairie voles tolerate a range of ambient temperatures, including conventional 22° housing, which makes them translationally appropriate for studies of diet-induced obesity. We also studied vole offspring for their growth, sucrose preference, home cage locomotor activity, and food consumption after birth by either cesarean section or vaginal delivery. At sacrifice, we collected measures of weight, length, and adipose tissue to analyze body composition in adulthood. Voles delivered by cesarean section had consistently greater bodyweights than those born vaginally, despite having lower food consumption and greater locomotive activity. Cesarean-delivered animals were also longer, though this did not explain their greater body weights. While cesarean delivery had no effect on vasopressin, it resulted in less oxytocin immunoreactivity within the hypothalamus in adulthood. These results support the case that cesarean section delivery plays a causal role in increasing offspring body weight, potentially by affecting the oxytocin system.
Collapse
Affiliation(s)
- William M Kenkel
- Department of Psychological and Brain Sciences, University of Delaware, United States of America.
| | - Sabreen Ahmed
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Miranda Partie
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| | - Katelyn Rogers
- Department of Psychological and Brain Sciences, University of Delaware, United States of America
| |
Collapse
|
7
|
Mauvais-Jarvis F. Sex differences in energy metabolism: natural selection, mechanisms and consequences. Nat Rev Nephrol 2024; 20:56-69. [PMID: 37923858 DOI: 10.1038/s41581-023-00781-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/06/2023]
Abstract
Metabolic homeostasis operates differently in men and women. This sex asymmetry is the result of evolutionary adaptations that enable women to resist loss of energy stores and protein mass while remaining fertile in times of energy deficit. During starvation or prolonged exercise, women rely on oxidation of lipids, which are a more efficient energy source than carbohydrates, to preserve glucose for neuronal and placental function and spare proteins necessary for organ function. Carbohydrate reliance in men could be an evolutionary adaptation related to defence and hunting, as glucose, unlike lipids, can be used as a fuel for anaerobic high-exertion muscle activity. The larger subcutaneous adipose tissue depots in healthy women than in healthy men provide a mechanism for lipid storage. As female mitochondria have higher functional capacity and greater resistance to oxidative damage than male mitochondria, uniparental inheritance of female mitochondria may reduce the transmission of metabolic disorders. However, in women, starvation resistance and propensity to obesity have evolved in tandem, and the current prevalence of obesity is greater in women than in men. The combination of genetic sex, programming by developmental testosterone in males, and pubertal sex hormones defines sex-specific biological systems in adults that produce phenotypic sex differences in energy homeostasis, metabolic disease and drug responses.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine and Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA.
- Endocrine service, Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
| |
Collapse
|
8
|
Alarcon PC, Damen MSMA, Ulanowicz CJ, Sawada K, Oates JR, Toth A, Wayland JL, Chung H, Stankiewicz TE, Moreno-Fernandez ME, Szabo S, Zacharias WJ, Divanovic S. Obesity amplifies influenza virus-driven disease severity in male and female mice. Mucosal Immunol 2023; 16:843-858. [PMID: 37730122 PMCID: PMC10842771 DOI: 10.1016/j.mucimm.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Influenza virus-induced respiratory pneumonia remains a major public health concern. Obesity, metabolic diseases, and female sex are viewed as independent risk factors for worsened influenza virus-induced lung disease severity. However, lack of experimental models of severe obesity in female mice limits discovery-based studies. Here, via utility of thermoneutral housing (30 °C) and high-fat diet (HFD) feeding, we induced severe obesity and metabolic disease in female C57BL/6 mice and compared their responses to severely obese male C57BL/6 counterparts during influenza virus infection. We show that lean male and female mice have similar lung edema, inflammation, and immune cell infiltration during influenza virus infection. At standard housing conditions, HFD-fed male, but not female, mice exhibit severe obesity, metabolic disease, and exacerbated influenza disease severity. However, combining thermoneutral housing and HFD feeding in female mice induces severe obesity and metabolic disease, which is sufficient to amplify influenza virus-driven disease severity to a level comparable to severely obese male counterparts. Lastly, increased total body weights of male and female mice at time of infection correlated with worsened influenza virus-driven disease severity metrics. Together, our findings confirm the impact of obesity and metabolic disease as key risk factors to influenza disease severity and present a novel mouse experimental model suitable for future mechanistic interrogation of sex, obesity, and metabolic disease traits in influenza virus-driven disease severity.
Collapse
Affiliation(s)
- Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Cassidy J Ulanowicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrea Toth
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jennifer L Wayland
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Gastroenterology, Hepatology and Nutrition Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sara Szabo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - William J Zacharias
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
9
|
Wayland JL, Doll JR, Lawson MJ, Stankiewicz TE, Oates JR, Sawada K, Damen MSMA, Alarcon PC, Haslam DB, Trout AT, DeFranco EA, Klepper CM, Woo JG, Moreno-Fernandez ME, Mouzaki M, Divanovic S. Thermoneutral Housing Enables Studies of Vertical Transmission of Obesogenic Diet-Driven Metabolic Diseases. Nutrients 2023; 15:4958. [PMID: 38068816 PMCID: PMC10708424 DOI: 10.3390/nu15234958] [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: 10/28/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Vertical transmission of obesity is a critical contributor to the unabated obesity pandemic and the associated surge in metabolic diseases. Existing experimental models insufficiently recapitulate "human-like" obesity phenotypes, limiting the discovery of how severe obesity in pregnancy instructs vertical transmission of obesity. Here, via utility of thermoneutral housing and obesogenic diet feeding coupled to syngeneic mating of WT obese female and lean male mice on a C57BL/6 background, we present a tractable, more "human-like" approach to specifically investigate how maternal obesity contributes to offspring health. Using this model, we found that maternal obesity decreased neonatal survival, increased offspring adiposity, and accelerated offspring predisposition to obesity and metabolic disease. We also show that severe maternal obesity was sufficient to skew offspring microbiome and create a proinflammatory gestational environment that correlated with inflammatory changes in the offspring in utero and adulthood. Analysis of a human birth cohort study of mothers with and without obesity and their infants was consistent with mouse study findings of maternal inflammation and offspring weight gain propensity. Together, our results show that dietary induction of obesity in female mice coupled to thermoneutral housing can be used for future mechanistic interrogations of obesity and metabolic disease in pregnancy and vertical transmission of pathogenic traits.
Collapse
Affiliation(s)
- Jennifer L. Wayland
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jessica R. Doll
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Matthew J. Lawson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Traci E. Stankiewicz
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jarren R. Oates
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Keisuke Sawada
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle S. M. A. Damen
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pablo C. Alarcon
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - David B. Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Andrew T. Trout
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Department of Radiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Emily A. DeFranco
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Corie M. Klepper
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jessica G. Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maria E. Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Marialena Mouzaki
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Senad Divanovic
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
Walendzik K, Kopcewicz M, Wiśniewska J, Opyd P, Machcińska-Zielińska S, Gawrońska-Kozak B. Dermal white adipose tissue development and metabolism: The role of transcription factor Foxn1. FASEB J 2023; 37:e23171. [PMID: 37682531 DOI: 10.1096/fj.202300873rr] [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: 05/02/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Intradermal adipocytes form dermal white adipose tissue (dWAT), a unique fat depot localized in the lower layer of the dermis. However, recognition of molecular factors regulating dWAT development, homeostasis, and bioactivity is limited. Using Foxn1-/- and Foxn1+/+ mice, we demonstrated that epidermally expressed Foxn1 regulates dWAT development and defines the adipogenic capacity of dermal fibroblasts. In intact and post-wounded skin, Foxn1 contributes to the initial stimulation of dWAT adipogenesis and participates in the modulation of lipid metabolism processes. Furthermore, Foxn1 activity strengthens adipogenic processes through Bmp2 and Igf2 signaling and regulates lipid metabolism in differentiated dermal fibroblasts. The results reveal the contribution of Foxn1 to dWAT metabolism, thus identifying possible targets for modulation and regulation of dWAT in physiological and pathological processes in the skin.
Collapse
Affiliation(s)
- Katarzyna Walendzik
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Wiśniewska
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Paulina Opyd
- Department of Animal Nutrition and Feed Science, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Sylwia Machcińska-Zielińska
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawrońska-Kozak
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
11
|
Hylander BL, Qiao G, Cortes Gomez E, Singh P, Repasky EA. Housing temperature plays a critical role in determining gut microbiome composition in research mice: Implications for experimental reproducibility. Biochimie 2023; 210:71-81. [PMID: 36693616 PMCID: PMC10953156 DOI: 10.1016/j.biochi.2023.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Preclinical mouse models are widely used for studying mechanisms of disease and responses to therapeutics, however there is concern about the lack of experimental reproducibility and failure to predict translational success. The gut microbiome has emerged as a regulator of metabolism and immunological processes in health and disease. The gut microbiome of mice differs by supplier and this affects experimental outcomes. We have previously reported that the mandated, mildly cool housing temperature for research mice (22°-26 °C) induces chronic adrenergic stress which suppresses anti-tumor immunity and promotes tumor growth compared to thermoneutral housing (30 °C). Therefore, we wondered how housing temperature affects the microbiome. Here, we demonstrate that the gut microbiome of BALB/c mice is easily modulated by a few degrees difference in temperature. Our results reveal significant differences between the gut microbiome of mice housed at 22°-23 °C vs. 30 °C. Although the genera vary, we consistently observed an enrichment of members of the family Lachnospiraceae when mice are housed at 22°-23 °C. These findings demonstrate that adrenergic stress and need for increased energy harvest to support thermogenesis, in addition to other factors such as diet, modulates the gut microbiome and this could be one mechanism by which housing temperature affects experimental outcomes. Additionally, tumor growth in mice housed at 30 °C also increases the proportion of Lachnospiraceae. The idea that stress can alter the gut microbiome and cause differences in experimental outcomes is applicable to mouse studies in general and is a variable that has significant potential to affect experimental reproducibility.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Guanxi Qiao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Eduardo Cortes Gomez
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Prashant Singh
- Genomics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
12
|
Queen NJ, Huang W, Komatineni S, Mansour AG, Xiao R, Chrislip LA, Cao L. Social isolation exacerbates diet-induced obesity and peripheral inflammation in young male mice under thermoneutrality. iScience 2023; 26:106259. [PMID: 36915694 PMCID: PMC10006833 DOI: 10.1016/j.isci.2023.106259] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/10/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Social isolation (SI) is associated with an increased risk of mortality and various chronic diseases-including obesity-in humans. Murine studies probing SI metabolic outcomes remain inconsistent, due in part to a lack of consideration for housing temperature. Such experiments typically occur at room temperature, subjecting mice to chronic cold stress. Single housing prevents social thermoregulation, further exacerbating cold stress and obscuring psychosocial influences on metabolism at room temperature. In this study, C57BL/6 and BALB/c male mice were group- and single-housed under thermoneutral conditions to determine whether SI affects the development of high-fat diet-induced obesity. We report SI promotes weight gain, increases food intake, increases adiposity, worsens glycemic control, reduces insulin signaling, exacerbates systemic and adipose inflammatory responses, and induces a molecular signature within the hypothalamus. This study establishes a murine model that recapitulates the SI-induced propensity for obesity, which may further our understanding of SI's influence on health and disease.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Wei Huang
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Suraj Komatineni
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Anthony G. Mansour
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope, National Medical Center and the Beckman Research Institute, Los Angeles, CA 91010, USA
| | - Run Xiao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Logan A. Chrislip
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
13
|
Zhang BT, Xu JY, Wang W, Zeng Y, Jiang J. Obesity and cancer: Mouse models used in studies. Front Oncol 2023; 13:1125178. [PMID: 37007087 PMCID: PMC10061215 DOI: 10.3389/fonc.2023.1125178] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
There is increasing evidence that obesity is associated with the occurrence and development of malignant tumors. When studying the relationship between obesity and malignant tumors, it is very important to choose an appropriate animal model. However, BALB/c nude mice and other animals commonly used to study tumor xenograft (human-derived tumor cell lines) transplantation models are difficult to induce obesity, while C57BL/6 mice and other model animals commonly used for obesity research are not suitable for tumor xenograft transplantation. Therefore, it is difficult to replicate both obesity and malignancy in animal models at the same time. This review summarizes several experimental animal models and protocols that can simultaneously induce obesity and tumor xenografts.
Collapse
Affiliation(s)
- Bo-Tao Zhang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia-Ying Xu
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Wang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Zeng
- Department of Orthodontic, the Affiliated Stomatological Hospital of Southwest Medical University, Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
- *Correspondence: Jun Jiang, ; Yang Zeng,
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Jun Jiang, ; Yang Zeng,
| |
Collapse
|
14
|
Ginting RP, Lee JM, Lee MW. The Influence of Ambient Temperature on Adipose Tissue Homeostasis, Metabolic Diseases and Cancers. Cells 2023; 12:cells12060881. [PMID: 36980222 PMCID: PMC10047443 DOI: 10.3390/cells12060881] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Adipose tissue is a recognized energy storage organ during excessive energy intake and an endocrine and thermoregulator, which interacts with other tissues to regulate systemic metabolism. Adipose tissue dysfunction is observed in most obese mouse models and humans. However, most studies using mouse models were conducted at room temperature (RT), where mice were chronically exposed to mild cold. In this condition, energy use is prioritized for thermogenesis to maintain body temperature in mice. It also leads to the activation of the sympathetic nervous system, followed by the activation of β-adrenergic signaling. As humans live primarily in their thermoneutral (TN) zone, RT housing for mice limits the interpretation of disease studies from mouse models to humans. Therefore, housing mice in their TN zone (~28–30 °C) can be considered to mimic humans physiologically. However, factors such as temperature ranges and TN pre-acclimatization periods should be examined to obtain reliable results. In this review, we discuss how adipose tissue responds to housing temperature and the outcomes of the TN zone in metabolic disease studies. This review highlights the critical role of TN housing in mouse models for studying adipose tissue function and human metabolic diseases.
Collapse
Affiliation(s)
- Rehna Paula Ginting
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Ji-Min Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Min-Woo Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
- Correspondence: ; Tel.: +82-41-413-5029
| |
Collapse
|
15
|
Lac M, Tavernier G, Moro C. Does housing temperature influence glucose regulation and muscle-fat crosstalk in mice? Biochimie 2023:S0300-9084(23)00028-7. [PMID: 36758717 DOI: 10.1016/j.biochi.2023.01.019] [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: 08/18/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
The robustness of scientific results is partly based on their reproducibility. Working with animal models, particularly in the field of metabolism, requires to avoid any source of stress to rule out a maximum of bias. Housing at room temperature is sufficient to induce thermal stress activating key thermogenic organs such as brown adipose tissue (BAT) and skeletal muscle. BAT covers most of the non-shivering thermogenesis in mice and burns a variety of fuels such as glucose and lipids. A high prevalence of BAT is associated with a strong protection against type 2 diabetes risk in humans, implying that BAT plays a key role in glucose homeostasis. However, thermal stress is poorly and inconsistently considered in experimental research. This thermal stress can significantly impede interpretation of phenotypes by favoring compensatory signaling pathways. Indeed, various studies revealed that thermoneutrality is essential to study metabolism in mice in order to reach a suitable level of "humanization". In this review, we briefly discuss if and how ambient temperature influence blood glucose homeostasis through BAT and muscle-fat crosstalk.
Collapse
Affiliation(s)
- Marlène Lac
- Institute of Metabolic and Cardiovascular Diseases, Team MetaDiab, INSERM, Paul Sabatier University, UMR1297, Toulouse, France
| | - Geneviève Tavernier
- Institute of Metabolic and Cardiovascular Diseases, Team MetaDiab, INSERM, Paul Sabatier University, UMR1297, Toulouse, France
| | - Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, Team MetaDiab, INSERM, Paul Sabatier University, UMR1297, Toulouse, France.
| |
Collapse
|
16
|
Tognolli K, Silva V, Sousa-Filho CPB, Cardoso CAL, Gorjão R, Otton R. Green tea beneficial effects involve changes in the profile of immune cells in the adipose tissue of obese mice. Eur J Nutr 2023; 62:321-336. [PMID: 35994086 DOI: 10.1007/s00394-022-02963-3] [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: 10/11/2021] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE During obesity, the adipose tissue is usually infiltrated by immune cells which are related to hallmarks of obesity such as systemic inflammation and insulin resistance (IR). Green tea (GT) has been widely studied for its anti-inflammatory actions, including the modulation in the proliferation and activity of immune cells, in addition to preventing cardiovascular and metabolic diseases. METHODS The aim of the present study was to analyze the population of immune cells present in the subcutaneous and epididymal white adipose tissue (WAT) of mice kept at thermoneutrality (TN) and fed with a high-fat diet (HFD) for 16 weeks, supplemented or not with GT extract (500 mg/kg/12 weeks). RESULTS The HFD in association with TN has induced chronic inflammation, and IR in parallel with changes in the profile of immune cells in the subcutaneous and epidydimal WAT, increasing pro-inflammatory cytokines release, inflammatory cells infiltration, and fibrotic aspects in WAT. On the other hand, GT prevented body weight gain, in addition to avoiding IR and inflammation, and the consequent tissue fibrosis, maintaining a lower concentration of cytokines and a profile of immune cells similar to the control mice, preventing the harmful modulations induced by both HFD and TN. CONCLUSIONS GT beneficial effects in WAT abrogated the deleterious effects triggered by HFD and TN, maintaining all immune cells and fibrotic markers at the same level as in lean mice. These results place WAT immune cells population as a potential target of GT action, also highlighting the positive effects of GT in obese mice housed at TN.
Collapse
Affiliation(s)
- Kaue Tognolli
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Victoria Silva
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Celso Pereira Batista Sousa-Filho
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | | | - Renata Gorjão
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Rosemari Otton
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil.
| |
Collapse
|
17
|
Keller AC, Chun JH, Knaub L, Henckel M, Hull S, Scalzo R, Pott G, Walker L, Reusch J. Thermoneutrality induces vascular dysfunction and impaired metabolic function in male Wistar rats: a new model of vascular disease. J Hypertens 2022; 40:2133-2146. [PMID: 35881464 PMCID: PMC9553250 DOI: 10.1097/hjh.0000000000003153] [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: 10/26/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Cardiovascular disease is of paramount importance, yet there are few relevant rat models to investigate its pathology and explore potential therapeutics. Housing at thermoneutral temperature (30 °C) is being employed to humanize metabolic derangements in rodents. We hypothesized that housing rats in thermoneutral conditions would potentiate a high-fat diet, resulting in diabetes and dysmetabolism, and deleteriously impact vascular function, in comparison to traditional room temperature housing (22 °C). METHODS Male Wistar rats were housed at either room temperature or thermoneutral temperatures for 16 weeks on either a low or high-fat diet. Glucose and insulin tolerance tests were conducted at the beginning and end of the study. At the study's conclusion, vasoreactivity and mitochondrial respiration of aorta and carotid were conducted. RESULTS We observed diminished vasodilation in vessels from thermoneutral rats ( P < 0.05), whereas high-fat diet had no effect. This effect was also observed in endothelium-denuded aorta in thermoneutral rats ( P < 0.05). Vasoconstriction was significantly elevated in aorta of thermoneutral rats ( P < 0.05). Diminished nitric oxide synthase activity and nitrotyrosine, and elevated glutathione activity were observed in aorta from rats housed under thermoneutral conditions, indicating a climate of lower nitric oxide and excess reactive oxygen species in aorta. Thermoneutral rat aorta also demonstrated less mitochondrial respiration with lipid substrates compared with the controls ( P < 0.05). CONCLUSION Our data support that thermoneutrality causes dysfunctional vasoreactivity, decreased lipid mitochondrial metabolism, and modified cellular signaling. These are critical observations as thermoneutrality is becoming prevalent for translational research models. This new model of vascular dysfunction may be useful for dissection of targetable aspects of cardiovascular disease and is a novel and necessary model of disease.
Collapse
Affiliation(s)
- Amy C. Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | | | - L.A. Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - M.M. Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - S.E. Hull
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - R.L. Scalzo
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - G.B. Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - L.A. Walker
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - J.E.B. Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| |
Collapse
|
18
|
Phung HH, Lee CH. Mouse models of nonalcoholic steatohepatitis and their application to new drug development. Arch Pharm Res 2022; 45:761-794. [DOI: 10.1007/s12272-022-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
19
|
Kowaltowski AJ. Cold Exposure and the Metabolism of Mice, Men, and Other Wonderful Creatures. Physiology (Bethesda) 2022; 37:0. [PMID: 35575253 DOI: 10.1152/physiol.00002.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Laboratory rodents and cold-adapted animals in the wild use a significant amount of the energy derived from food intake for heat generation. Thermogenesis involving mitochondrial uncoupling in the brown adipose tissue differs quantitatively in mice, humans, and cold-adapted animals and could be an important ally to combat obesity if humans were prepared to deviate slightly from thermoneutral living conditions to activate this pathway.
Collapse
Affiliation(s)
- Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
21
|
Chun JH, Henckel MM, Knaub LA, Hull SE, Pott GB, Ramirez DG, Reusch JEB, Keller AC. (-)-Epicatechin Reverses Glucose Intolerance in Rats Housed at Thermoneutrality. PLANTA MEDICA 2022; 88:735-744. [PMID: 35777366 PMCID: PMC9343939 DOI: 10.1055/a-1843-9855] [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] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/04/2022] [Indexed: 06/09/2023]
Abstract
Diabetes is a life-threatening and debilitating disease with pathological hallmarks, including glucose intolerance and insulin resistance. Plant compounds are a source of novel and effective therapeutics, and the flavonoid (-)-epicatechin, common to popular foods worldwide, has been shown to improve carbohydrate metabolism in both clinical studies and preclinical models. We hypothesized that (-)-epicatechin would alleviate thermoneutral housing-induced glucose intolerance. Male rats were housed at either thermoneutral (30 °C) or room temperature (24 °C) for 16 weeks and gavaged with either 1 mg/kg body weight or vehicle for the last 15 days before sacrifice. Rats housed at thermoneutrality had a significantly elevated serum glucose area under the curve (p < 0.05) and reduced glucose-mediated insulin secretion. In contrast, rats at thermoneutrality treated with (-)-epicatechin had improved glucose tolerance and increased insulin secretion (p < 0.05). Insulin tolerance tests revealed no differences in insulin sensitivity in any of the four groups. Pancreatic immunohistochemistry staining showed significantly greater islet insulin positive cells in animals housed at thermoneutrality. In conclusion, (-)-epicatechin improved carbohydrate tolerance via increased insulin secretion in response to glucose challenge without a change in insulin sensitivity.
Collapse
Affiliation(s)
- Ji Hye Chun
- Aquillius Corp., San Diego, CA, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Melissa M. Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Leslie A. Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Sara E. Hull
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Greg B. Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - David G. Ramirez
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Jane E.-B. Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Amy C. Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| |
Collapse
|
22
|
James CM, Olejniczak SH, Repasky EA. How murine models of human disease and immunity are influenced by housing temperature and mild thermal stress. Temperature (Austin) 2022; 10:166-178. [PMID: 37332306 PMCID: PMC10274546 DOI: 10.1080/23328940.2022.2093561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022] Open
Abstract
At the direction of The Guide and Use of Laboratory Animals, rodents in laboratory facilities are housed at ambient temperatures between 20°C and 26°C, which fall below their thermoneutral zone (TNZ). TNZ is identified as a range of ambient temperatures that allow an organism to regulate body temperature without employing additional thermoregulatory processes (e.g. metabolic heat production driven by norepinephrine), thus leading to mild, chronic cold stress. For mice, this chronic cold stress leads to increased serum levels of the catecholamine norepinephrine, which has direct effects on various immune cells and several aspects of immunity and inflammation. Here, we review several studies that have revealed that ambient temperature significantly impacts outcomes in various murine models of human diseases, particularly those in which the immune system plays a major role in its pathogenesis. The impact of ambient temperature on experimental outcomes raises questions regarding the clinical relevance of some murine models of human disease, since studies examining rodents housed within thermoneutral ambient temperatures revealed that rodent disease pathology more closely resembled that of humans. Unlike laboratory rodents, humans can modify their surroundings accordingly - by adjusting their clothing, the thermostat, or their physical activity - to live within the appropriate TNZ, offering a possible explanation for why many studies using murine models of human disease conducted at thermoneutrality better represent patient outcomes. Thus, it is strongly recommended that ambient housing temperature in such studies be consistently and accurately reported and recognized as an important experimental variable.
Collapse
Affiliation(s)
- Caitlin M. James
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | |
Collapse
|
23
|
Hamilton A, Rizzo R, Brod S, Ono M, Perretti M, Cooper D, D'Acquisto F. The immunomodulatory effects of social isolation in mice are linked to temperature control. Brain Behav Immun 2022; 102:179-194. [PMID: 35217174 DOI: 10.1016/j.bbi.2022.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/17/2022] [Accepted: 02/18/2022] [Indexed: 12/25/2022] Open
Abstract
Living in isolation is considered an emerging societal problem that negatively affects the physical wellbeing of its sufferers in ways that we are just starting to appreciate. This study investigates the immunomodulatory effects of social isolation in mice, utilising a two-week program of sole cage occupancy followed by the testing of immune-inflammatory resilience to bacterial sepsis. Our results revealed that mice housed in social isolation showed an increased ability to clear bacterial infection compared to control socially housed animals. These effects were associated with specific changes in whole blood gene expression profile and an increased production of classical pro-inflammatory cytokines. Interestingly, equipping socially isolated mice with artificial nests as a substitute for their natural huddling behaviour reversed the increased resistance to bacterial sepsis. Together these results suggest that the control of body temperature through social housing and huddling behaviour are important factors in the regulation of the host immune response to infection in mice and might provide another example of the many ways by which living conditions influence immunity.
Collapse
Affiliation(s)
- Alice Hamilton
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Raffaella Rizzo
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Samuel Brod
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Masahiro Ono
- University of London Imperial College Science Technology & Medicine, Department of Life Science, Faculty of Natural Science, London SW7 2AZ, England
| | - Mauro Perretti
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dianne Cooper
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Fulvio D'Acquisto
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; School of Life and Health Science, University of Roehampton, London SW15, 4JD, UK.
| |
Collapse
|
24
|
Molekularne mechanizmy działania czynnika transkrypcyjnego FOXN1 w skórze. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstrakt
Artykuł jest przeglądem wyników badań dotyczących funkcji i mechanizmu działania czynnika transkrypcyjnego FOXN1. Lokalizacja FOXN1 u przedstawicieli wszystkich ssaków ogranicza się do nabłonka tylko dwóch organów: skóry i grasicy. W skórze FOXN1 stymuluje różnicowanie się keratynocytów, reguluje proces pigmentacji i bierze udział w rozwoju włosów. W skórze objętej urazem FOXN1 jest zaangażowany w bliznowy proces gojenia poprzez udział w reepitelializacji oraz w procesie przejścia epitelialno-mezenchymalnego (epithelial-mesenchymal transition; EMT). Pozbawione aktywnego czynnika transkrypcyjnego FOXN1 dorosłe myszy (Foxn1-/-) goją urazy skórne w unikalnym, charakterystycznym jedynie dla płodów ssaków, procesie bezbliznowej (scar-free) regeneracji. Analiza porównawcza transkryptomów skóry: dorosłych myszy Foxn1-/- oraz skóry płodów myszy (14. dzień rozwoju płodowego) wykazała istotne podobieństwa w ekspresji genów związanych przede wszystkim z przebudową tkanek, budową cytoszkieletu, gojeniem urazów, odpowiedzią immunologiczną oraz różnicowaniem. Wyniki te wskazują, iż FOXN1 może być głównym elementem szlaku sygnałowego na drodze tzw. punktu tranzycyjnego czyli przejścia z etapu gojenia bezbliznowego (płodowego) do bliznowego (dorosłego) w trakcie rozwoju płodowego.
Collapse
|
25
|
Glenny EM, Coleman MF, Giles ED, Wellberg EA, Hursting SD. Designing Relevant Preclinical Rodent Models for Studying Links Between Nutrition, Obesity, Metabolism, and Cancer. Annu Rev Nutr 2021; 41:253-282. [PMID: 34357792 PMCID: PMC8900211 DOI: 10.1146/annurev-nutr-120420-032437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diet and nutrition are intricately related to cancer prevention, growth, and treatment response. Preclinical rodent models are a cornerstone to biomedical research and remain instrumental in our understanding of the relationship between cancer and diet and in the development of effective therapeutics. However, the success rate of translating promising findings from the bench to the bedside is suboptimal. Well-designed rodent models will be crucial to improving the impact basic science has on clinical treatment options. This review discusses essential experimental factors to consider when designing a preclinical cancer model with an emphasis on incorporatingthese models into studies interrogating diet, nutrition, and metabolism. The aims of this review are to (a) provide insight into relevant considerations when designing cancer models for obesity, nutrition, and metabolism research; (b) identify common pitfalls when selecting a rodent model; and (c) discuss strengths and limitations of available preclinical models.
Collapse
Affiliation(s)
- Elaine M Glenny
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Michael F Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Erin D Giles
- Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, USA
| |
Collapse
|
26
|
Seeley RJ, MacDougald OA. Mice as experimental models for human physiology: when several degrees in housing temperature matter. Nat Metab 2021; 3:443-445. [PMID: 33767444 PMCID: PMC8987294 DOI: 10.1038/s42255-021-00372-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Standfirst: Some “species differences” between mouse and human can be diminished simply by housing mice at warmer temperatures. Failure to strategically turn up the thermostat may undermine translation of findings in mice into insights on human metabolic diseases.
Collapse
Affiliation(s)
- Randy J Seeley
- Departments of Surgery, Internal Medicine and Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA.
| | - Ormond A MacDougald
- Departments of Molecular & Integrative Physiology, and Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
27
|
Tan PY, Teng KT. Role of dietary fat on obesity-related postmenopausal breast cancer: insights from mouse models and methodological considerations. Breast Cancer 2021; 28:556-571. [PMID: 33687609 DOI: 10.1007/s12282-021-01233-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/23/2021] [Indexed: 01/02/2023]
Abstract
The increasing incidence rate of breast cancer in the last few decades is known to be linked to the upward trend of obesity prevalence worldwide. The consumption of high-fat diet in particular has been correlated with postmenopausal breast cancer risk. The underlying mechanisms, using suitable and reliable experimental mouse model, however, is lacking. The current review aims to discuss the evidence available from mouse models on the effects of dietary fats intake on postmenopausal breast cancer. We will further discuss the biochemical mechanisms involved in the occurrence of postmenopausal breast cancer. In addition, the methodological considerations and their limitations in obesity-related postmenopausal breast cancer, such as choice of mouse models and breast cancer cell lines as well as the study duration will be reviewed. The current review will provide a platform for further development of new xenograft models which may offer the opportunity to investigate the mechanisms of postmenopausal breast cancer in a greater detail.
Collapse
Affiliation(s)
- Pei Yee Tan
- Division of Product Development and Advisory Services, Malaysian Palm Oil Board, No 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Kim Tiu Teng
- Division of Product Development and Advisory Services, Malaysian Palm Oil Board, No 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia.
| |
Collapse
|
28
|
Giles ED, Wellberg EA. Preclinical Models to Study Obesity and Breast Cancer in Females: Considerations, Caveats, and Tools. J Mammary Gland Biol Neoplasia 2020; 25:237-253. [PMID: 33146844 PMCID: PMC8197449 DOI: 10.1007/s10911-020-09463-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/09/2020] [Indexed: 12/28/2022] Open
Abstract
Obesity increases the risk for breast cancer and is associated with poor outcomes for cancer patients. A variety of rodent models have been used to investigate these relationships; however, key differences in experimental approaches, as well as unique aspects of rodent physiology lead to variability in how these valuable models are implemented. We combine expertise in the development and implementation of preclinical models of obesity and breast cancer to disseminate effective practices for studies that integrate these fields. In this review, we share, based on our experience, key considerations for model selection, highlighting important technical nuances and tips for use of preclinical models in studies that integrate obesity with breast cancer risk and progression. We describe relevant mouse and rat paradigms, specifically highlighting differences in breast tumor subtypes, estrogen production, and strategies to manipulate hormone levels. We also outline options for diet composition and housing environments to promote obesity in female rodents. While we have applied our experience to understanding obesity-associated breast cancer, the experimental variables we incorporate have relevance to multiple fields that investigate women's health.
Collapse
Affiliation(s)
- Erin D Giles
- Department of Nutrition, Texas A&M University, College Station, TX, USA.
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| |
Collapse
|
29
|
Vialard F, Olivier M. Thermoneutrality and Immunity: How Does Cold Stress Affect Disease? Front Immunol 2020; 11:588387. [PMID: 33329571 PMCID: PMC7714907 DOI: 10.3389/fimmu.2020.588387] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
One of the major challenges the scientific community faces today is the lack of translational data generated from mouse trials for human health application. Housing temperature-dependent chronic cold stress in laboratory rodents is one of the key factors contributing to lack of translatability because it reveals major metabolic differences between humans and rodents. While humans tend to operate at temperatures within their thermoneutral zone, most laboratory rodents are housed at temperatures below this zone and have an increased energy demand to generate heat. This has an impact on the immune system of mice and thus affects results obtained using murine models of human diseases. A limited number of studies and reviews have shown that results obtained on mice housed at thermoneutrality were different from those obtained from mice housed in traditional housing conditions. Most of those studies, focused on obesity and cancer, found that housing mice at thermoneutrality changed the outcomes of the diseases negatively and positively, respectively. In this review, we describe how thermoneutrality impacts the immune system of rodents generally and in the context of different disease models. We show that thermoneutrality exacerbates cardiovascular and auto-immune diseases; alleviates asthma and Alzheimer’s disease; and, changes gut microbiome populations. We also show that thermoneutrality can have exacerbating or alleviating effects on the outcome of infectious diseases. Thus, we join the call of others in this field to urge researchers to refine murine models of disease and increase their translational capacity by considering housing at thermoneutrality for trials involving rodents.
Collapse
Affiliation(s)
- Fiorella Vialard
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Lee VK, David JM, Huerkamp MJ. Micro- and Macroenvironmental Conditions and Stability of Terrestrial Models. ILAR J 2020; 60:120-140. [PMID: 33094820 DOI: 10.1093/ilar/ilaa013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 01/15/2023] Open
Abstract
Environmental variables can have profound effects on the biological responses of research animals and the outcomes of experiments dependent on them. Some of these influences are both predictable and unpredictable in effect, many are challenging to standardize, and all are influenced by the planning and conduct of experiments and the design and operation of the vivarium. Others are not yet known. Within the immediate environment where the research animal resides, in the vivarium and in transit, the most notable of these factors are ambient temperature, relative humidity, gaseous pollutant by-products of animal metabolism and physiology, dust and particulates, barometric pressure, electromagnetic fields, and illumination. Ambient temperatures in the animal housing environment, in particular those experienced by rodents below the thermoneutral zone, may introduce degrees of stress and thermoregulatory compensative responses that may complicate or invalidate study measurements across a broad array of disciplines. Other factors may have more subtle and specific effects. It is incumbent on scientists designing and executing experiments and staff responsible for animal husbandry to be aware of, understand, measure, systematically record, control, and account for the impact of these factors on sensitive animal model systems to ensure the quality and reproducibility of scientific studies.
Collapse
Affiliation(s)
- Vanessa K Lee
- Department of Pathology and Laboratory Medicine and Division of Animal Resources, School of Medicine, Emory University, Atlanta, Georgia
| | - John M David
- Translational Medicine Department, Vertex Pharmaceuticals, Boston, Massachusetts
| | - Michael J Huerkamp
- Department of Pathology and Laboratory Medicine and Division of Animal Resources, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
31
|
Enriquez J, Mims BMD, Trasti S, Furr KL, Grisham MB. Genomic, microbial and environmental standardization in animal experimentation limiting immunological discovery. BMC Immunol 2020; 21:50. [PMID: 32878597 PMCID: PMC7464063 DOI: 10.1186/s12865-020-00380-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background The use of inbred mice housed under standardized environmental conditions has been critical in identifying immuno-pathological mechanisms in different infectious and inflammatory diseases as well as revealing new therapeutic targets for clinical trials. Unfortunately, only a small percentage of preclinical intervention studies using well-defined mouse models of disease have progressed to clinically-effective treatments in patients. The reasons for this lack of bench-to-bedside transition are not completely understood; however, emerging data suggest that genetic diversity and housing environment may greatly influence muring immunity and inflammation. Results Accumulating evidence suggests that certain immune responses and/or disease phenotypes observed in inbred mice may be quite different than those observed in their outbred counterparts. These differences have been thought to contribute to differing immune responses to foreign and/or auto-antigens in mice vs. humans. There is also a growing literature demonstrating that mice housed under specific pathogen free conditions possess an immature immune system that remarkably affects their ability to respond to pathogens and/or inflammation when compared with mice exposed to a more diverse spectrum of microorganisms. Furthermore, recent studies demonstrate that mice develop chronic cold stress when housed at standard animal care facility temperatures (i.e. 22–24 °C). These temperatures have been shown alter immune responses to foreign and auto-antigens when compared with mice housed at their thermo-neutral body temperature of 30–32 °C. Conclusions Exposure of genetically diverse mice to a spectrum of environmentally-relevant microorganisms at housing temperatures that approximate their thermo-neutral zone may improve the chances of identifying new and more potent therapeutics to treat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Josue Enriquez
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Brianyell Mc Daniel Mims
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Scott Trasti
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.,Laboratory Animal Research Center, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Kathryn L Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Matthew B Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.
| |
Collapse
|
32
|
Turner RT, Philbrick KA, Wong CP, Gamboa AR, Branscum AJ, Iwaniec UT. Effects of Propranolol on Bone, White Adipose Tissue, and Bone Marrow Adipose Tissue in Mice Housed at Room Temperature or Thermoneutral Temperature. Front Endocrinol (Lausanne) 2020; 11:117. [PMID: 32256446 PMCID: PMC7089918 DOI: 10.3389/fendo.2020.00117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
Growing female mice housed at room temperature (22°C) weigh the same but differ in body composition compared to mice housed at thermoneutrality (32°C). Specifically, mice housed at room temperature have lower levels of white adipose tissue (WAT). Additionally, bone marrow adipose tissue (bMAT) and cancellous bone volume fraction in distal femur metaphysis are lower in room temperature-housed mice. The metabolic changes induced by sub-thermoneutral housing are associated with lower leptin levels in serum and higher levels of Ucp1 gene expression in brown adipose tissue. Although the precise mechanisms mediating adaptation to sub-thermoneutral temperature stress remain to be elucidated, there is evidence that increased sympathetic nervous system activity acting via β-adrenergic receptors plays an important role. We therefore evaluated the effect of the non-specific β-blocker propranolol (primarily β1 and β2 antagonist) on body composition, femur microarchitecture, and bMAT in growing female C57BL/6 mice housed at either room temperature or thermoneutral temperature. As anticipated, cancellous bone volume fraction, WAT and bMAT were lower in mice housed at room temperature. Propranolol had small but significant effects on bone microarchitecture (increased trabecular number and decreased trabecular spacing), but did not attenuate premature bone loss induced by room temperature housing. In contrast, propranolol treatment prevented housing temperature-associated differences in WAT and bMAT. To gain additional insight, we evaluated a panel of genes in tibia, using an adipogenesis PCR array. Housing temperature and treatment with propranolol had exclusive as well as shared effects on gene expression. Of particular interest was the finding that room temperature housing reduced, whereas propranolol increased, expression of the gene for acetyl-CoA carboxylase (Acacb), the rate-limiting step for fatty acid synthesis and a key regulator of β-oxidation. Taken together, these findings provide evidence that increased activation of β1 and/or β2 receptors contributes to reduced bMAT by regulating adipocyte metabolism, but that this pathway is unlikely to be responsible for premature cancellous bone loss in room temperature-housed mice.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, United States
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Carmen P. Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Amanda R. Gamboa
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, United States
- *Correspondence: Urszula T. Iwaniec
| |
Collapse
|
33
|
Walendzik K, Kopcewicz M, Bukowska J, Panasiewicz G, Szafranska B, Gawronska-Kozak B. The Transcription Factor FOXN1 Regulates Skin Adipogenesis and Affects Susceptibility to Diet-Induced Obesity. J Invest Dermatol 2019; 140:1166-1175.e9. [PMID: 31811821 DOI: 10.1016/j.jid.2019.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Abstract
FOXN1, a transcription factor expressed in the epidermis, regulates keratinocyte differentiation and participates in skin wound healing. In this study, we explored the impact of FOXN1 insufficiency on diet-stimulated weight gain and dermal white adipose tissue regulation in the intact and wounded skin of FOXN1eGFP/+ (heterozygotes, FOXN1-insufficient) mice in the context of age and diet. The results showed that on a high-fat diet, FOXN1eGFP/+ mice gained significantly less body weight than their FOXN1+/+ counterparts (FOXN1-sufficient mice). The intact and wounded skin of FOXN1eGFP/+ mice displayed abrogated expression of the master regulators of adipogenesis, PPARγ, FABP4, and leptin, which decreased with age in FOXN1+/+ mice. FOXN1 insufficiency also resulted in a decreased percentage of adipocyte-committed precursor cells (CD24+) in the skin. The proadipogenic pathway genes Bmp2, Igf2, and Mest showed a gradual decrease in expression that accompanied the gradual inactivation of FOXN1 in the skin of FOXN1+/+, FOXN1eGFP/+, and FOXN1eGFP/eGFP (lack of FOXN1) mice. Bone morphogenetic protein 2 and insulin-like growth factor 2 signals colocalized with FOXN1-eGFP in the epidermis and in hair follicles. These data demonstrated that FOXN1 initiates the cascade of adipogenic signaling that regulates skin homeostasis and wound healing and affects susceptibility to diet-induced obesity.
Collapse
Affiliation(s)
- Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Bukowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Grzegorz Panasiewicz
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Bozena Szafranska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.
| |
Collapse
|
34
|
Martin SA, Philbrick KA, Wong CP, Olson DA, Branscum AJ, Jump DB, Marik CK, DenHerder JM, Sargent JL, Turner RT, Iwaniec UT. Thermoneutral housing attenuates premature cancellous bone loss in male C57BL/6J mice. Endocr Connect 2019; 8:1455-1467. [PMID: 31590144 PMCID: PMC6865368 DOI: 10.1530/ec-19-0359] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
Mice are a commonly used model to investigate aging-related bone loss but, in contrast to humans, mice exhibit cancellous bone loss prior to skeletal maturity. The mechanisms mediating premature bone loss are not well established. However, our previous work in female mice suggests housing temperature is a critical factor. Premature cancellous bone loss was prevented in female C57BL/6J mice by housing the animals at thermoneutral temperature (where basal rate of energy production is at equilibrium with heat loss). In the present study, we determined if the protective effects of thermoneutral housing extend to males. Male C57BL/6J mice were housed at standard room temperature (22°C) or thermoneutral (32°C) conditions from 5 (rapidly growing) to 16 (slowly growing) weeks of age. Mice housed at room temperature exhibited reductions in cancellous bone volume fraction in distal femur metaphysis and fifth lumbar vertebra; these effects were abolished at thermoneutral conditions. Mice housed at thermoneutral temperature had higher levels of bone formation in distal femur (based on histomorphometry) and globally (serum osteocalcin), and lower global levels of bone resorption (serum C-terminal telopeptide of type I collagen) compared to mice housed at room temperature. Thermoneutral housing had no impact on bone marrow adiposity but resulted in higher abdominal white adipose tissue and serum leptin. The overall magnitude of room temperature housing-induced cancellous bone loss did not differ between male (current study) and female (published data) mice. These findings highlight housing temperature as a critical experimental variable in studies using mice of either sex to investigate aging-related changes in bone metabolism.
Collapse
Affiliation(s)
- Stephen A Martin
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Kenneth A Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Carmen P Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Dawn A Olson
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Adam J Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Charles K Marik
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Jonathan M DenHerder
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Jennifer L Sargent
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon, USA
- Correspondence should be addressed to U T Iwaniec:
| |
Collapse
|
35
|
Hylander BL, Gordon CJ, Repasky EA. Manipulation of Ambient Housing Temperature To Study the Impact of Chronic Stress on Immunity and Cancer in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:631-636. [PMID: 30670578 DOI: 10.4049/jimmunol.1800621] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
Mice are the preeminent research organism in which to model human diseases and study the involvement of the immune response. Rapidly accumulating evidence indicates a significant involvement of stress hormones in cancer progression, resistance to therapies, and suppression of immune responses. As a result, there has been a concerted effort to model human stress in mice. In this article, we discuss recent literature showing how mice in research facilities are chronically stressed at baseline because of environmental factors. Focusing on housing temperature, we suggest that the stress of cool housing temperatures contributes to the impact of other imposed experimental stressors and therefore has a confounding effect on mouse stress models. Furthermore, we propose that manipulation of housing temperature is a useful approach for studying the impact of chronic stress on disease and the immune response and for testing therapeutic methods of reducing the negative effects of chronic stress.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Christopher J Gordon
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| |
Collapse
|
36
|
Oxidation Stress-Mediated MAPK Signaling Pathway Activation Induces Neuronal Loss in the CA1 and CA3 Regions of the Hippocampus of Mice Following Chronic Cold Exposure. Brain Sci 2019; 9:brainsci9100273. [PMID: 31614701 PMCID: PMC6826747 DOI: 10.3390/brainsci9100273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic stress can damage homeostasis and induce various primary diseases. Although chronic cold stress is becoming an increasing problem for people who must work or live in extreme environments, risk-induced diseases in the central nervous system remain unstudied. Male C57BL/6 mice were exposed to an environment of 4 °C, 3 h per day for 1, 2, and 3 weeks and homeostasis in the hippocampus and neuronal apoptosis were evaluated by Western blotting, immunohistochemistry, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and immunofluorescence. The phenomena of oxidation stress, MAPK signaling pathway activation, anti-oxidation protein release, neuronal apoptosis increases, and neuronal proliferation inhibition were demonstrated in the CA1 and CA3 regions of mouse hippocampal tissues following cold exposure. We speculated that these phenomena were mediated by the MAPK pathway and were closely linked with oxidative stress in the hippocampus. This study provides novel concepts regarding neurodegenerative diseases, suggesting that chronic cold stress may be a critical factor to induce neurodegenerative diseases.
Collapse
|
37
|
Liu Q, Zhou Z, Liu P, Zhang S. Comparative proteomic study of liver lipid droplets and mitochondria in mice housed at different temperatures. FEBS Lett 2019; 593:2118-2138. [PMID: 31234227 PMCID: PMC6771624 DOI: 10.1002/1873-3468.13509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 01/18/2023]
Abstract
Laboratory mice are standardly housed at around 23 °C, setting them under chronic cold stress. Metabolic changes in the liver in mice housed at thermoneutral, standard and cold temperatures remain unknown. In the present study, we isolated lipid droplets and mitochondria from their livers in a comparative proteomic study aiming to investigate the changes. According to proteomic analysis, mitochondrial tricarboxylic acid cycle (TCA cycle) and retinol metabolism are enhanced, whereas oxidative phosphorylation is not affected obviously under cold conditions, suggesting that liver mitochondria may increase TCA cycle capacity in biosynthetic pathways, as well as retinol metabolism, to help the liver to adapt. Based on proteomic and immunoblotting results, perilipin 5 and major urinary proteins are increased significantly, whereas mitochondrial pyruvate carrier is decreased dramatically under cold conditions, indicating their involvement in liver adaptation.
Collapse
Affiliation(s)
- Qingfeng Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ziyun Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingsheng Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
38
|
Albustanji L, Perez GS, AlHarethi E, Aldiss P, Bloor I, Barreto-Medeiros JM, Budge H, Symonds ME, Dellschaft N. Housing Temperature Modulates the Impact of Diet-Induced Rise in Fat Mass on Adipose Tissue Before and During Pregnancy in Rats. Front Physiol 2019; 10:209. [PMID: 30894820 PMCID: PMC6414463 DOI: 10.3389/fphys.2019.00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/18/2019] [Indexed: 11/13/2022] Open
Abstract
Aim: To investigate whether housing temperature influences rat adiposity, and the extent it is modified by diet and/or pregnancy. Housing temperature impacts on brown adipose tissue, that possess a unique uncoupling protein (UCP) 1, which, when activated by reduced ambient temperature, enables rapid heat generation. Methods: We, therefore, examined whether the effects of dietary induced rise in fat mass on interscapular brown fat in female rats were dependent on housing temperature, and whether pregnancy further modulates the response. Four week old rats were either maintained at thermoneutrality (27°C) or at a “standard” cool temperature (20°C), and fed either a control or obesogenic (high in fat and sugar) diet until 10 weeks old. They were then either tissue sampled or mated with a male maintained under the same conditions. The remaining dams were tissue sampled at either 10 or 19 days gestation. Results: Diet had the greatest effect on fat mass at thermoneutrality although, by 19 days gestation, fat weight was similar between groups. Prior to mating, the abundance of UCP1 was higher at 20°C, but was similar between groups during pregnancy. UCP1 mRNA followed a similar pattern, with expression declining to a greater extent in the animals maintained at 20°C. Conclusion: Housing temperature has a marked influence on the effect of dietary induced rise in fat deposition that was modified through gestation. This maybe mediated by the reduction in UCP1 with housing at thermoneutrality prior to pregnancy and could subsequently impact on growth and development of the offspring.
Collapse
Affiliation(s)
- Layla Albustanji
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Gabriela S Perez
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom.,Graduate Program of Food Nutrition and Health, Department of Food Science, School of Nutrition, Federal University of Bahia, Salvador, Brazil.,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Enas AlHarethi
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Peter Aldiss
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Ian Bloor
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Jairza M Barreto-Medeiros
- Graduate Program of Food Nutrition and Health, Department of Food Science, School of Nutrition, Federal University of Bahia, Salvador, Brazil.,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Helen Budge
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom.,Nottingham Digestive Disease Centre and Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Neele Dellschaft
- Early Life Research Unit, Division of Child Health, Obstetrics, and Gynaecology, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
39
|
Small L, Gong H, Yassmin C, Cooney GJ, Brandon AE. Thermoneutral housing does not influence fat mass or glucose homeostasis in C57BL/6 mice. J Endocrinol 2018; 239:313-324. [PMID: 30400016 DOI: 10.1530/joe-18-0279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022]
Abstract
One major factor affecting physiology often overlooked when comparing data from animal models and humans is the effect of ambient temperature. The majority of rodent housing is maintained at ~22°C, the thermoneutral temperature for lightly clothed humans. However, mice have a much higher thermoneutral temperature of ~30°C, consequently data collected at 22°C in mice could be influenced by animals being exposed to a chronic cold stress. The aim of this study was to investigate the effect of housing temperature on glucose homeostasis and energy metabolism of mice fed normal chow or a high-fat, obesogenic diet (HFD). Male C57BL/6J(Arc) mice were housed at standard temperature (22°C) or at thermoneutrality (29°C) and fed either chow or a 60% HFD for 13 weeks. The HFD increased fat mass and produced glucose intolerance as expected but this was not exacerbated in mice housed at thermoneutrality. Changing the ambient temperature, however, did alter energy expenditure, food intake, lipid content and glucose metabolism in skeletal muscle, liver and brown adipose tissue. Collectively, these findings demonstrate that mice regulate energy balance at different housing temperatures to maintain whole-body glucose tolerance and adiposity irrespective of the diet. Despite this, metabolic differences in individual tissues were apparent. In conclusion, dietary intervention in mice has a greater impact on adiposity and glucose metabolism than housing temperature although temperature is still a significant factor in regulating metabolic parameters in individual tissues.
Collapse
Affiliation(s)
- Lewin Small
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
| | - Henry Gong
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Christian Yassmin
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Zhang C, Seo J, Murakami K, Salem ESB, Bernhard E, Borra VJ, Choi K, Yuan CL, Chan CC, Chen X, Huang T, Weirauch MT, Divanovic S, Qi NR, Thomas HE, Mercer CA, Siomi H, Nakamura T. Hepatic Ago2-mediated RNA silencing controls energy metabolism linked to AMPK activation and obesity-associated pathophysiology. Nat Commun 2018; 9:3658. [PMID: 30201950 PMCID: PMC6131149 DOI: 10.1038/s41467-018-05870-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
RNA silencing inhibits mRNA translation. While mRNA translation accounts for the majority of cellular energy expenditure, it is unclear if RNA silencing regulates energy homeostasis. Here, we report that hepatic Argonaute 2 (Ago2)-mediated RNA silencing regulates both intrinsic energy production and consumption and disturbs energy metabolism in the pathogenesis of obesity. Ago2 regulates expression of specific miRNAs including miR-802, miR-103/107, and miR-148a/152, causing metabolic disruption, while simultaneously suppressing the expression of genes regulating glucose and lipid metabolism, including Hnf1β, Cav1, and Ampka1. Liver-specific Ago2-deletion enhances mitochondrial oxidation and ATP consumption associated with mRNA translation, which results in AMPK activation, and improves obesity-associated pathophysiology. Notably, hepatic Ago2-deficiency improves glucose metabolism in conditions of insulin receptor antagonist treatment, high-fat diet challenge, and hepatic AMPKα1-deletion. The regulation of energy metabolism by Ago2 provides a novel paradigm in which RNA silencing plays an integral role in determining basal metabolic activity in obesity-associated sequelae.
Collapse
Affiliation(s)
- Cai Zhang
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Joonbae Seo
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kazutoshi Murakami
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Esam S B Salem
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elise Bernhard
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vishnupriya J Borra
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Celvie L Yuan
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Calvin C Chan
- Medical Scientist Training Program, Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Xiaoting Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Taosheng Huang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nathan R Qi
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hala Einakat Thomas
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Carol A Mercer
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Haruhiko Siomi
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, Japan
| | - Takahisa Nakamura
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
41
|
Wellberg EA, Kabos P, Gillen AE, Jacobsen BM, Brechbuhl HM, Johnson SJ, Rudolph MC, Edgerton SM, Thor AD, Anderson SM, Elias A, Zhou XK, Iyengar NM, Morrow M, Falcone DJ, El-Hely O, Dannenberg AJ, Sartorius CA, MacLean PS. FGFR1 underlies obesity-associated progression of estrogen receptor-positive breast cancer after estrogen deprivation. JCI Insight 2018; 3:120594. [PMID: 30046001 PMCID: PMC6124402 DOI: 10.1172/jci.insight.120594] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/19/2018] [Indexed: 01/07/2023] Open
Abstract
Obesity increases breast cancer mortality by promoting resistance to therapy. Here, we identified regulatory pathways in estrogen receptor-positive (ER-positive) tumors that were shared between patients with obesity and those with resistance to neoadjuvant aromatase inhibition. Among these was fibroblast growth factor receptor 1 (FGFR1), a known mediator of endocrine therapy resistance. In a preclinical model with patient-derived ER-positive tumors, diet-induced obesity promoted a similar gene expression signature and sustained the growth of FGFR1-overexpressing tumors after estrogen deprivation. Tumor FGFR1 phosphorylation was elevated with obesity and predicted a shorter disease-free and disease-specific survival for patients treated with tamoxifen. In both human and mouse mammary adipose tissue, FGF1 ligand expression was associated with metabolic dysfunction, weight gain, and adipocyte hypertrophy, implicating the impaired response to a positive energy balance in growth factor production within the tumor niche. In conjunction with these studies, we describe a potentially novel graft-competent model that can be used with patient-derived tissue to elucidate factors specific to extrinsic (host) and intrinsic (tumor) tissue that are critical for obesity-associated tumor promotion. Taken together, we demonstrate that obesity and excess energy establish a tumor environment with features of endocrine therapy resistance and identify a role for ligand-dependent FGFR1 signaling in obesity-associated breast cancer progression.
Collapse
Affiliation(s)
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Austin E. Gillen
- University of Colorado School of Medicine, RNA Bioscience Initiative, Aurora, Colorado, USA
| | - Britta M. Jacobsen
- Department of Pathology and
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Heather M. Brechbuhl
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Michael C. Rudolph
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | - Anthony Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York, USA
| | - Neil M. Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Monica Morrow
- Department of Surgery, MSKCC, New York, New York, USA
| | - Domenick J. Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Omar El-Hely
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | | | - Paul S. MacLean
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
42
|
Polyethylene particles inserted over calvarium induce cancellous bone loss in femur in female mice. Bone Rep 2018; 9:84-92. [PMID: 30094298 PMCID: PMC6073052 DOI: 10.1016/j.bonr.2018.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/13/2018] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
Focal bone resorption (osteolysis) induced by wear particles contributes to long-term orthopedic joint failure. However, the impact of focal osteolysis on remote skeletal sites has received less attention. The goal of this study was to determine the effects of polyethylene particles placed over calvaria on representative axial and appendicular skeletal sites in female mice. Because recent work has identified housing temperature as an important biological variable in mice, response to particle treatment was measured in animals housed at room (22 °C) and thermoneutral (32 °C) temperature. Osteolysis was evident in skeletal tissue adjacent to particle insertion. In addition, cancellous bone loss was observed in distal femur metaphysis. The bone loss was associated with lower osteoblast-lined perimeter and lower mineralizing perimeter in distal femur, lower osteocalcin gene expression in tibia, and lower serum osteocalcin, suggesting the response was due, at least in part, to reduced bone formation. Mild cold stress induced by sub-thermoneutral housing resulted in cancellous bone loss in distal femur and lumbar vertebra but did not influence skeletal response to particles. In summary, the results indicate that focal inflammation induced by polyethylene particles has the potential to result in systemic bone loss. This is significant because bone loss is a risk factor for fracture. Wear particles contribute to orthopedic joint failure by promoting focal inflammation- mediated osteolysis. Here we investigated effects of polyethylene particles placed over calvaria on remote skeletal sites in female mice. Particles placed over calvaria resulted in focal inflammation and cancellous bone loss in distal femur metaphysis. Lower osteoblast-lined perimeter suggests reduced bone formation contributed to bone loss in distal femur. Our results suggest focal inflammation induced by polyethylene particles has the potential to result in systemic bone loss.
Collapse
|
43
|
Foxn1 in Skin Development, Homeostasis and Wound Healing. Int J Mol Sci 2018; 19:ijms19071956. [PMID: 29973508 PMCID: PMC6073674 DOI: 10.3390/ijms19071956] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/29/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023] Open
Abstract
Intensive research effort has focused on cellular and molecular mechanisms that regulate skin biology, including the phenomenon of scar-free skin healing during foetal life. Transcription factors are the key molecules that tune gene expression and either promote or suppress gene transcription. The epidermis is the source of transcription factors that regulate many functions of epidermal cells such as proliferation, differentiation, apoptosis, and migration. Furthermore, the activation of epidermal transcription factors also causes changes in the dermal compartment of the skin. This review focuses on the transcription factor Foxn1 and its role in skin biology. The regulatory function of Foxn1 in the skin relates to physiological (development and homeostasis) and pathological (skin wound healing) conditions. In particular, the pivotal role of Foxn1 in skin development and the acquisition of the adult skin phenotype, which coincides with losing the ability of scar-free healing, is discussed. Thus, genetic manipulations with Foxn1 expression, specifically those introducing conditional Foxn1 silencing in a Foxn1+/+ organism or its knock-in in a Foxn1−/− model, may provide future perspectives for regenerative medicine.
Collapse
|
44
|
Clayton ZS, McCurdy CE. Short-term thermoneutral housing alters glucose metabolism and markers of adipose tissue browning in response to a high-fat diet in lean mice. Am J Physiol Regul Integr Comp Physiol 2018; 315:R627-R637. [PMID: 29791203 DOI: 10.1152/ajpregu.00364.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Systemic insulin resistance and glucose intolerance occur with as little as 3 days of a high-fat diet (HFD) in mice and humans; the mechanisms that initiate acute insulin resistance are unknown. Most laboratories house mice at 22°C, which is below their thermoneutral temperature (~30°C). Cold stress has been shown to increase white adipose tissue (WAT) browning, alter lipid trafficking, and impair immune function, whereas energy intake and expenditure decrease with increasing ambient temperature; importantly, dysregulation of these parameters has been strongly linked to obesity-induced insulin resistance. Therefore, we compared acute changes in glucose metabolism and the metabolic phenotype in lean mice in response to a control diet or HFD housed at standard vivarium (22°C) and thermoneutral (30°C) temperatures. Glucose intolerance occurred following 1 or 5 days of HFD and was independent of housing temperature or adiposity; however, the reduction in tissue-specific glucose clearance with HFD diverged by temperature with reduced brown adipose tissue (BAT) glucose uptake at 22°C but reduced soleus glucose uptake at 30°C. Fasting glucose, food intake, and energy expenditure were significantly lower at 30°C, independent of diet. Additionally, markers of browning in both BAT and inguinal subcutaneous WAT, but not perigonadal epididymal WAT, decreased at 30°C. Together, we find housing temperature has a significant impact on the cellular pathways that regulate glucose tolerance in response to an acute HFD exposure. Thus, even short-term changes in housing temperature should be highly considered in interpretation of metabolic studies in mice.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Human Physiology, University of Oregon , Eugene, Oregon
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon , Eugene, Oregon
| |
Collapse
|
45
|
Moreno-Fernandez ME, Giles DA, Stankiewicz TE, Sheridan R, Karns R, Cappelletti M, Lampe K, Mukherjee R, Sina C, Sallese A, Bridges JP, Hogan SP, Aronow BJ, Hoebe K, Divanovic S. Peroxisomal β-oxidation regulates whole body metabolism, inflammatory vigor, and pathogenesis of nonalcoholic fatty liver disease. JCI Insight 2018; 3:93626. [PMID: 29563328 DOI: 10.1172/jci.insight.93626] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), a metabolic predisposition for development of hepatocellular carcinoma (HCC), represents a disease spectrum ranging from steatosis to steatohepatitis to cirrhosis. Acox1, a rate-limiting enzyme in peroxisomal fatty acid β-oxidation, regulates metabolism, spontaneous hepatic steatosis, and hepatocellular damage over time. However, it is unknown whether Acox1 modulates inflammation relevant to NAFLD pathogenesis or if Acox1-associated metabolic and inflammatory derangements uncover and accelerate potential for NAFLD progression. Here, we show that mice with a point mutation in Acox1 (Acox1Lampe1) exhibited altered cellular metabolism, modified T cell polarization, and exacerbated immune cell inflammatory potential. Further, in context of a brief obesogenic diet stress, NAFLD progression associated with Acox1 mutation resulted in significantly accelerated and exacerbated hepatocellular damage via induction of profound histological changes in hepatocytes, hepatic inflammation, and robust upregulation of gene expression associated with HCC development. Collectively, these data demonstrate that β-oxidation links metabolism and immune responsiveness and that a better understanding of peroxisomal β-oxidation may allow for discovery of mechanisms central for NAFLD progression.
Collapse
Affiliation(s)
- Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Daniel A Giles
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA.,Immunology Graduate Program, CCHMC, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Rachel Sheridan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Pathology, CCHMC, Cincinnati, Ohio, USA
| | - Rebekah Karns
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Gastroenterology, Hepatology, and Nutrition, CCHMC, Cincinnati, Ohio, USA
| | - Monica Cappelletti
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Kristin Lampe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Rajib Mukherjee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Christian Sina
- Molecular Gastroenterology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Anthony Sallese
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Neonatology and Pulmonary Biology
| | - James P Bridges
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Neonatology and Pulmonary Biology
| | - Simon P Hogan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Allergy and Immunology, and
| | - Bruce J Aronow
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Biomedical Informatics, CCHMC, Cincinnati, Ohio, USA
| | - Kasper Hoebe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| |
Collapse
|
46
|
Rodriguez-Palacios A, Aladyshkina N, Ezeji JC, Erkkila HL, Conger M, Ward J, Webster J, Cominelli F. 'Cyclical Bias' in Microbiome Research Revealed by A Portable Germ-Free Housing System Using Nested Isolation. Sci Rep 2018; 8:3801. [PMID: 29491439 PMCID: PMC5830500 DOI: 10.1038/s41598-018-20742-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/12/2018] [Indexed: 12/24/2022] Open
Abstract
Germ-Free (GF) research has required highly technical pressurized HEPA-ventilation anchored systems for decades. Herein, we validated a GF system that can be easily implemented and portable using Nested Isolation (NesTiso). GF-standards can be achieved housing mice in non-HEPA-static cages, which only need to be nested 'one-cage-inside-another' resembling 'Russian dolls'. After 2 years of monitoring ~100,000 GF-mouse-days, NesTiso showed mice can be maintained GF for life (>1.3 years), with low animal daily-contamination-probability risk (1 every 867 days), allowing the expansion of GF research with unprecedented freedom and mobility. At the cage level, with 23,360 GF cage-days, the probability of having a cage contamination in NesTiso cages opened in biosafety hoods was statistically identical to that of opening cages inside (the 'gold standard') multi-cage pressurized GF isolators. When validating the benefits of using NesTiso in mouse microbiome research, our experiments unexpectedly revealed that the mouse fecal microbiota composition within the 'bedding material' of conventional SPF-cages suffers cyclical selection bias as moist/feces/diet/organic content ('soiledness') increases over time (e.g., favoring microbiome abundances of Bacillales, Burkholderiales, Pseudomonadales; and cultivable Enterococcus faecalis over Lactobacillus murinus and Escherichia coli), which in turn cyclically influences the gut microbiome dynamics of caged mice. Culture 'co-streaking' assays showed that cohoused mice exhibiting different fecal microbiota/hemolytic profiles in clean bedding (high-within-cage individual diversity) 'cyclically and transiently appear identical' (less diverse) as bedding soiledness increases, and recurs. Strategies are proposed to minimize this novel functional form of cyclical bedding-dependent microbiome selection bias.
Collapse
Affiliation(s)
- Alexander Rodriguez-Palacios
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Natalia Aladyshkina
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jessica C Ezeji
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Hailey L Erkkila
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Mathew Conger
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - John Ward
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Joshua Webster
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| |
Collapse
|
47
|
Thermoneutral housing temperature regulates T-regulatory cell function and inhibits ovabumin-induced asthma development in mice. Sci Rep 2017; 7:7123. [PMID: 28769099 PMCID: PMC5540912 DOI: 10.1038/s41598-017-07471-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
The change in ambient temperature is one of the risk factors for the aggravation of bronchial asthma (BA). Yet, whether the ambient temperature influences the immune functions associated with allergic asthma remains unknown. In this study, we treated asthmatic mice with standard temperature (ST, 20 °C) or thermoneutral temperature (TT, 30 °C). The results showed that the airway inflammatory cell counts in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) were significantly reduced in the mice treated with TT as compared with the mice treated with ST. The imbalance of Th1/Th2 response in the lung was improved following housing the mice at TT. In addition, the pulmonary Treg cells were increased in asthmatic mice after TT treatment. The temperature stress (29 °C and 41 °C) drove naïve CD4T cells towards Th2 cells. Our data demonstrate that the change of ambient temperature was a risk factor to aggravate experimental asthma.
Collapse
|
48
|
Giles DA, Moreno-Fernandez ME, Stankiewicz TE, Graspeuntner S, Cappelletti M, Wu D, Mukherjee R, Chan CC, Lawson MJ, Klarquist J, Sünderhauf A, Softic S, Kahn CR, Stemmer K, Iwakura Y, Aronow BJ, Karns R, Steinbrecher KA, Karp CL, Sheridan R, Shanmukhappa SK, Reynaud D, Haslam DB, Sina C, Rupp J, Hogan SP, Divanovic S. Thermoneutral housing exacerbates nonalcoholic fatty liver disease in mice and allows for sex-independent disease modeling. Nat Med 2017; 23:829-838. [PMID: 28604704 PMCID: PMC5596511 DOI: 10.1038/nm.4346] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/22/2017] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a common prelude to cirrhosis and hepatocellular carcinoma, is the most common chronic liver disease worldwide. Defining the molecular mechanisms underlying the pathogenesis of NAFLD has been hampered by a lack of animal models that closely recapitulate the severe end of the disease spectrum in humans, including bridging hepatic fibrosis. Here we demonstrate that a novel experimental model employing thermoneutral housing, as opposed to standard housing, resulted in lower stress-driven production of corticosterone, augmented mouse proinflammatory immune responses and markedly exacerbated high-fat diet (HFD)-induced NAFLD pathogenesis. Disease exacerbation at thermoneutrality was conserved across multiple mouse strains and was associated with augmented intestinal permeability, an altered microbiome and activation of inflammatory pathways that are associated with the disease in humans. Depletion of Gram-negative microbiota, hematopoietic cell deletion of Toll-like receptor 4 (TLR4) and inactivation of the IL-17 axis resulted in altered immune responsiveness and protection from thermoneutral-housing-driven NAFLD amplification. Finally, female mice, typically resistant to HFD-induced obesity and NAFLD, develop full disease characteristics at thermoneutrality. Thus, thermoneutral housing provides a sex-independent model of exacerbated NAFLD in mice and represents a novel approach for interrogation of the cellular and molecular mechanisms underlying disease pathogenesis.
Collapse
Affiliation(s)
- Daniel A Giles
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Monica Cappelletti
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Calvin C Chan
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Matthew J Lawson
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jared Klarquist
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Annika Sünderhauf
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Samir Softic
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebekah Karns
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Rachel Sheridan
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shiva K Shanmukhappa
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David B Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christian Sina
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
49
|
van den Berg SM, van Dam AD, Rensen PCN, de Winther MPJ, Lutgens E. Immune Modulation of Brown(ing) Adipose Tissue in Obesity. Endocr Rev 2017; 38:46-68. [PMID: 27849358 DOI: 10.1210/er.2016-1066] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Obesity is associated with a variety of medical conditions such as type 2 diabetes and cardiovascular diseases and is therefore responsible for high morbidity and mortality rates. Increasing energy expenditure by brown adipose tissue (BAT) is a current novel strategy to reduce the excessive energy stores in obesity. Brown adipocytes burn energy to generate heat and are mainly activated upon cold exposure. As prolonged cold exposure is not a realistic therapy, researchers worldwide are searching for novel ways to activate BAT and/or induce beiging of white adipose tissue. Recently, the contribution of immune cells in the regulation of brown adipocyte activity and beiging of white adipose tissue has gained increased attention, with a prominent role for eosinophils and alternatively activated macrophages. This review discusses the rediscovery of BAT, presents an overview of modes of activation and differentiation of beige and brown adipocytes, and describes the recently discovered immunological pathways that are key in mediating brown/beige adipocyte development and function. Interventions in immunological pathways harbor the potential to provide novel strategies to increase beige and brown adipose tissue activity as a therapeutic target for obesity.
Collapse
Affiliation(s)
- Susan M van den Berg
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands
| | - Andrea D van Dam
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Menno P J de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| |
Collapse
|
50
|
Hylander BL, Eng JWL, Repasky EA. The Impact of Housing Temperature-Induced Chronic Stress on Preclinical Mouse Tumor Models and Therapeutic Responses: An Important Role for the Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:173-189. [PMID: 29275472 PMCID: PMC9423006 DOI: 10.1007/978-3-319-67577-0_12] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the last 10-15 years, there has been a recognition that the catecholamines (norepinephrine, NE, and epinephrine, Epi) released by the sympathetic nervous system under stressful conditions promote tumor growth through a variety of mechanisms. Tumors recruit autonomic nerves during their development and NE is then released locally in the tumor microenvironment (TME). Acting through adrenergic receptors present on a variety of cells in the TME, NE and Epi induce proliferation, resistance to apoptosis, epithelial to mesenchymal transition, metastasis of tumor cells, angiogenesis, and inflammation in the TME. These pre-clinical studies have been conducted in mouse models whose care and housing parameters are outlined in "The Guide for the Care and Use of Laboratory Animals [1]. In particular, the Guide mandates that mice be housed at standardized sub-thermoneutral temperatures; however, this causes a state of chronic cold-stress and elevated levels of NE. Although mice are able to maintain a normal body temperature when kept at these cool temperatures, it is becoming clear that this cold-stress is sufficient to activate physiological changes which affect experimental outcomes. We find that when mice are housed under standard, sub-thermoneutral temperatures (~22 °C, ST), tumor growth is significantly greater than when mice are housed at thermoneutrality (~30 °C TT). We also find that the anti-tumor immune response is suppressed at ST and this immunosuppression can be reversed by housing mice at TT or by administration of propranolol (a β-adrenergic receptor antagonist) to mice housed at ST. Furthermore, at ST tumors are more resistant to therapy and can also be sensitized to cytotoxic therapies by housing mice at TT or by treating mice with propranolol. The implications of these observations are particularly relevant to the way in which experiments conducted in preclinical models are interpreted and the findings implemented in the clinic. It may be that the disappointing failure of many new therapies to fulfill their promise in the clinic is related to an incomplete preclinical assessment in mouse models. Further, an expanded understanding of the efficacy of a therapy alone or in combination obtained by testing under a wider range of conditions would better predict how patients, who are under various levels of stress, might respond in a clinical setting. This may be particularly important to consider since we now appreciate that long term outcome of many therapies depends on eliciting an immune response.It is clear that the outcome of metabolic experiments, immunological investigations and therapeutic efficacy testing in tumors of mice housed at ST is restricted and expanding these experiments to include results obtained at TT may provide us with valuable information that would otherwise be overlooked.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jason W-L Eng
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|