101
|
Smith LK, Arabi S, Lelliott EJ, McArthur GA, Sheppard KE. Obesity and the Impact on Cutaneous Melanoma: Friend or Foe? Cancers (Basel) 2020; 12:cancers12061583. [PMID: 32549336 PMCID: PMC7352630 DOI: 10.3390/cancers12061583] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Excess body weight has been identified as a risk factor for many types of cancers, and for the majority of cancers, it is associated with poor outcomes. In contrast, there are cancers in which obesity is associated with favorable outcomes and this has been termed the “obesity paradox”. In melanoma, the connection between obesity and the increased incidence is not as strong as for other cancer types with some but not all studies showing an association. However, several recent studies have indicated that increased body mass index (BMI) improves survival outcomes in targeted and immune therapy treated melanoma patients. The mechanisms underlying how obesity leads to changes in therapeutic outcomes are not completely understood. This review discusses the current evidence implicating obesity in melanoma progression and patient response to targeted and immunotherapy, and discusses potential mechanisms underpinning these associations.
Collapse
Affiliation(s)
- Lorey K. Smith
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (L.K.S.); (S.A.); (E.J.L.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shaghayegh Arabi
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (L.K.S.); (S.A.); (E.J.L.); (G.A.M.)
| | - Emily J. Lelliott
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (L.K.S.); (S.A.); (E.J.L.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Grant A. McArthur
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (L.K.S.); (S.A.); (E.J.L.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Karen E. Sheppard
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (L.K.S.); (S.A.); (E.J.L.); (G.A.M.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence:
| |
Collapse
|
102
|
Blagosklonny MV. From causes of aging to death from COVID-19. Aging (Albany NY) 2020; 12:10004-10021. [PMID: 32534452 PMCID: PMC7346074 DOI: 10.18632/aging.103493] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
COVID-19 is not deadly early in life, but mortality increases exponentially with age, which is the strongest predictor of mortality. Mortality is higher in men than in women, because men age faster, and it is especially high in patients with age-related diseases, such as diabetes and hypertension, because these diseases are manifestations of aging and a measure of biological age. At its deepest level, aging (a program-like continuation of developmental growth) is driven by inappropriately high cellular functioning. The hyperfunction theory of quasi-programmed aging explains why COVID-19 vulnerability (lethality) is an age-dependent syndrome, linking it to other age-related diseases. It also explains inflammaging and immunosenescence, hyperinflammation, hyperthrombosis, and cytokine storms, all of which are associated with COVID-19 vulnerability. Anti-aging interventions, such as rapamycin, may slow aging and age-related diseases, potentially decreasing COVID-19 vulnerability.
Collapse
|
103
|
Woodall MJ, Neumann S, Campbell K, Pattison ST, Young SL. The Effects of Obesity on Anti-Cancer Immunity and Cancer Immunotherapy. Cancers (Basel) 2020; 12:E1230. [PMID: 32422865 PMCID: PMC7281442 DOI: 10.3390/cancers12051230] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/29/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality worldwide. Traditional treatments include surgery, chemotherapy and radiation therapy, and more recently targeted therapies including immunotherapy are becoming routine care for some cancers. Immunotherapy aims to upregulate the patient's own immune system, enabling it to destroy cancerous cells. Obesity is a metabolic disorder characterized by significant weight that is an important contributor to many different diseases, including cancers. Obesity impacts the immune system and causes, among other things, a state of chronic low-grade inflammation. This is hypothesized to impact the efficacy of the immunotherapies. This review discusses the effects of obesity on the immune system and cancer immunotherapy, including the current evidence on the effect of obesity on immune checkpoint blockade, something which currently published reviews on this topic have not delved into. Data from several studies show that even though obesity causes a state of chronic low-grade inflammation with reductions in effector immune populations, it has a beneficial effect on patient survival following anti-PD-1/PD-L1 and anti-CTLA-4 treatment. However, research in this field is just emerging and further work is needed to expand our understanding of which cancer patients are likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Matthew J. Woodall
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (M.J.W.); (S.N.); (K.C.)
| | - Silke Neumann
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (M.J.W.); (S.N.); (K.C.)
| | - Katrin Campbell
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (M.J.W.); (S.N.); (K.C.)
| | - Sharon T. Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand;
| | - Sarah L. Young
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; (M.J.W.); (S.N.); (K.C.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
104
|
Turbitt WJ, Rosean CB, Weber KS, Norian LA. Obesity and CD8 T cell metabolism: Implications for anti-tumor immunity and cancer immunotherapy outcomes. Immunol Rev 2020; 295:203-219. [PMID: 32157710 PMCID: PMC7416819 DOI: 10.1111/imr.12849] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Obesity is an established risk factor for many cancers and has recently been found to alter the efficacy of T cell-based immunotherapies. Currently, however, the effects of obesity on immunometabolism remain unclear. Understanding these associations is critical, given the fact that T cell metabolism is tightly linked to effector function. Thus, any obesity-associated changes in T cell bioenergetics are likely to drive functional changes at the cellular level, alter the metabolome and cytokine/chemokine milieu, and impact cancer immunotherapy outcomes. Here, we provide a brief overview of T cell metabolism in the presence and absence of solid tumor growth and summarize current literature regarding obesity-associated changes in T cell function and bioenergetics. We also discuss recent findings related to the impact of host obesity on cancer immunotherapy outcomes and present potential mechanisms by which T cell metabolism may influence therapeutic efficacy. Finally, we describe promising pharmaceutical therapies that are being investigated for their ability to improve CD8 T cell metabolism and enhance cancer immunotherapy outcomes in patients, regardless of their obesity status.
Collapse
Affiliation(s)
- William J. Turbitt
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - K. Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah
| | - Lyse A. Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
105
|
Uremia-Associated Ageing of the Thymus and Adaptive Immune Responses. Toxins (Basel) 2020; 12:toxins12040224. [PMID: 32260178 PMCID: PMC7232426 DOI: 10.3390/toxins12040224] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
Progressive loss of renal function is associated with a series of changes of the adaptive immune system which collectively constitute premature immunological ageing. This phenomenon contributes significantly to the mortality and morbidity of end-stage renal disease (ESRD) patients. In this review, the effect of ESRD on the T cell part of the adaptive immune system is highlighted. Naïve T cell lymphopenia, in combination with the expansion of highly differentiated memory T cells, are the hallmarks of immunological ageing. The decreased production of newly formed T cells by the thymus is critically involved. This affects both the CD4 and CD8 T cell compartment and may contribute to the expansion of memory T cells. The expanding populations of memory T cells have a pro-inflammatory phenotype, add to low-grade inflammation already present in ESRD patients and destabilize atherosclerotic plaques. The effect of loss of renal function on the thymus is not reversed after restoring renal function by kidney transplantation and constitutes a long-term mortality risk factor. Promising results from animal experiments have shown that rejuvenation of the thymus is a possibility, although not yet applicable in humans.
Collapse
|
106
|
Tam BT, Morais JA, Santosa S. Obesity and ageing: Two sides of the same coin. Obes Rev 2020; 21:e12991. [PMID: 32020741 DOI: 10.1111/obr.12991] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/30/2019] [Indexed: 02/06/2023]
Abstract
Conditions and comorbidities of obesity mirror those of ageing and age-related diseases. Obesity and ageing share a similar spectrum of phenotypes such as compromised genomic integrity, impaired mitochondrial function, accumulation of intracellular macromolecules, weakened immunity, shifts in tissue and body composition, and enhanced systemic inflammation. Moreover, it has been shown that obesity reduces life expectancy by 5.8 years in men and 7.1 years in women after the age of 40. Shorter life expectancy could be because obesity holistically accelerates ageing at multiple levels. Besides jeopardizing nuclear DNA and mitochondrial DNA integrity, obesity modifies the DNA methylation pattern, which is associated with epigenetic ageing in different tissues. Additionally, other signs of ageing are seen in individuals with obesity including telomere shortening, systemic inflammation, and functional declines. This review aims to show how obesity and ageing are "two sides of the same coin" through discussing how obesity predisposes an individual to age-related conditions, illness, and disease. We will further demonstrate how the mechanisms that perpetuate the early-onset of chronic diseases in obesity parallel those of ageing.
Collapse
Affiliation(s)
- Bjorn T Tam
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Quebec, Montreal, Canada
| | - Jose A Morais
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Division of Geriatric Medicine and Research Institute, McGill University Health Centre, Quebec, Montreal, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Quebec, Montreal, Canada.,Research Centre, Centre intégré universitarie de santé et de services sociaux du Nord-de-I'Île-de-Montréal, Hôpital du Sacré-Cœur de Monréal (CIUSS-NIM, HSCM), Quebec, Montreal, Canada
| |
Collapse
|
107
|
JANECZKO-CZARNECKA MAŁGORZATA, RYBKA BLANKA, RYCZAN-KRAWCZYK RENATA, KAŁWAK KRZYSZTOF, USSOWICZ MAREK. Thymic activity in immune recovery after allogeneic hematopoietic stem cell transplantation in children. Cent Eur J Immunol 2020; 45:151-159. [PMID: 33456325 PMCID: PMC7792432 DOI: 10.5114/ceji.2019.89843] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Thymic output was studied prospectively in 52 children who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT). Thymic activity was assessed by quantification of recent thymic emigrants (RTE) discriminated from the rest of naive T cells by immunophenotype CD3+/CD4+/CD31+/CD45RA+. Thymic output was analyzed in correlation with the kinetics of immune recovery and in relation to other potential risk factors that may influence thymopoiesis: underlying disease, type of HSCT, source of stem cells, age of recipient and donor, type of conditioning, implemented graft versus host disease (GvHD) prophylaxis, viral reactivations (herpes viruses cytomegalovirus - CMV, Epstein-Barr virus - EBV, adenovirus - ADV, BK virus - BKV), occurrence and grade of both acute and chronic graft versus host disease (aGvHD, cGvHD) and number of transplanted CD34 cells/kg. The absolute count of RTE in peripheral blood was evaluated at 6 time points: before the conditioning and on days +15, +30, +60 , +90 and +180 after HSCT. Occurrence of grade II-IV aGvHD was the most important factor associated with low RTE counts after HSCT. History of malignant disease, and transplantation from matched unrelated donor were risk factors for lower thymic output. We found a weak inverse correlation between the age of the recipient and thymic output on post-HSCT day +180. Source of stem cells, type of conditioning, viral reactivations, occurrence of chronic GvHD, age of the donor and the number of transplanted CD34 cells/kg did not affect thymopoiesis in our study group. These preliminary findings and identification of risk factors for deterioration of thymic activity may in the future help in selecting candidates for thymus rejuvenation strategies.
Collapse
Affiliation(s)
- MAŁGORZATA JANECZKO-CZARNECKA
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - BLANKA RYBKA
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - RENATA RYCZAN-KRAWCZYK
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - KRZYSZTOF KAŁWAK
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - MAREK USSOWICZ
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
108
|
Exploratory examination of inflammation state, immune response and blood cell composition in a human obese cohort to identify potential markers predicting cancer risk. PLoS One 2020; 15:e0228633. [PMID: 32027700 PMCID: PMC7004330 DOI: 10.1371/journal.pone.0228633] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 01/21/2020] [Indexed: 01/12/2023] Open
Abstract
Obesity has reached epidemic proportions and is often accompanied by elevated levels of pro-inflammatory cytokines that promote many chronic diseases, including cancer. However, not all obese people develop these diseases and it would be very helpful to identify those at high risk early on so that preventative measures can be instituted. We performed an extensive evaluation of the effects of obesity on inflammatory markers, on innate and adaptive immune responses, and on blood cell composition to identify markers that might be useful in distinguishing those at elevated risk of cancer. Plasma samples from 42 volunteers with a BMI>35 had significantly higher CRP, PGE2, IL-1RA, IL-6 and IL-17 levels than 34 volunteers with normal BMIs. Of the cytokines and chemokines tested, only IL-17 was significantly higher in men with a BMI>35 than women with a BMI>35. As well, only IL-17 was significantly higher in those with a BMI>35 that had type 2 diabetes versus those without type 2 diabetes. Whole blood samples from participants with a BMI>35, when challenged with E. coli, produced significantly higher levels of IL-1RA while HSV-1 challenge resulted in significantly elevated IL-1RA and VEGF, and a non-significant increase in G-CSF and IL-8 levels. T cell activation of PBMCs, via anti-CD3 plus anti-CD28, resulted in significantly higher IFNγ production from volunteers with a BMI>35. In terms of blood cells, red blood cell distribution width (RDW), monocytes, granulocytes, CD4+T cells and Tregs were all significantly higher while, natural killer (NK) and CD8+ T cells were all significantly lower in the BMI>35 cohort, suggesting that obesity may reduce the ability to kill nascent tumor cells. Importantly, however, there was considerable person-to-person variation amongst participants with a BMI>35, with some volunteers showing markedly different values from controls and others showing normal levels of many parameters measured. These person-to-person variations may prove useful in identifying those at high risk of developing cancer.
Collapse
|
109
|
Dao MC, Saltzman E, Page M, Reece J, Mojtahed T, Wu D, Meydani SN. Lack of Differences in Inflammation and T Cell-Mediated Function between Young and Older Women with Obesity. Nutrients 2020; 12:E237. [PMID: 31963377 PMCID: PMC7019899 DOI: 10.3390/nu12010237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
Abstract
Both obesity and aging are associated with dysregulated immune and inflammatory responses. There is limited knowledge, however, on differences in the immune system between young and older adults with obesity. The goal of this study was to compare circulating inflammatory cytokines and T cell-mediated immune response between young and older women with obesity. Twenty-three young (23-43 years) and 21 older (60-83 years) women with obesity were recruited at the Weight and Wellness Center at Tufts Medical Center. Circulating inflammatory cytokines (CRP, IL-6, and IL-1β) and ex vivo indicators of T cell-mediated immune function were compared between the groups. Older women with obesity had significantly fewer circulating CD3+, CD8+, CD19+, and natural killer T (NKT) cells compared to young women with obesity (p = 0.016, p < 0.0001, p = 0.0003, and p < 0.0001, respectively). However, with few exceptions, there was no significant difference in inflammation markers or stimulated lymphocyte proliferation and cytokine production by peripheral blood mononuclear cells between young and older participants. These findings are in contrast to those previously reported in young and old subjects with healthy weight and call for further investigation into the impact of obesity on premature aging of the immune system.
Collapse
Affiliation(s)
- Maria Carlota Dao
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.S.); (T.M.); (D.W.); (S.N.M.)
| | - Edward Saltzman
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.S.); (T.M.); (D.W.); (S.N.M.)
| | - Melissa Page
- Weight and Wellness Center, Tufts Medical Center, Boston, MA 02111, USA; (M.P.); (J.R.)
| | - Jillian Reece
- Weight and Wellness Center, Tufts Medical Center, Boston, MA 02111, USA; (M.P.); (J.R.)
| | - Tara Mojtahed
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.S.); (T.M.); (D.W.); (S.N.M.)
| | - Dayong Wu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.S.); (T.M.); (D.W.); (S.N.M.)
| | - Simin Nikbin Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (E.S.); (T.M.); (D.W.); (S.N.M.)
| |
Collapse
|
110
|
de Frel DL, Atsma DE, Pijl H, Seidell JC, Leenen PJM, Dik WA, van Rossum EFC. The Impact of Obesity and Lifestyle on the Immune System and Susceptibility to Infections Such as COVID-19. Front Nutr 2020; 7:597600. [PMID: 33330597 PMCID: PMC7711810 DOI: 10.3389/fnut.2020.597600] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background: COVID-19 is a global challenge to healthcare. Obesity is common in patients with COVID-19 and seems to aggravate disease prognosis. In this review we explore the link between obesity, chronic disease, lifestyle factors and the immune system, and propose societal interventions to enhance global immunity. Search Strategy and Selection Criteria: We performed three literature searches using the keywords (1) coronavirus AND comorbidities, (2) comorbidities AND immune system, and (3) lifestyle factors AND immune system. Results were screened for relevance by the main author and a total of 215 articles were thoroughly analyzed. Results: The relationship between obesity and unfavorable COVID-19 prognosis is discussed in light of the impact of chronic disease and lifestyle on the immune system. Several modifiable lifestyle factors render us susceptible to viral infections. In this context, we make a case for fostering a healthy lifestyle on a global scale. Conclusions: Obesity, additional chronic disease and an unhealthy lifestyle interactively impair immune function and increase the risk of severe infectious disease. In adverse metabolic and endocrine conditions, the immune system is geared toward inflammation. Collective effort is needed to ameliorate modifiable risk factors for obesity and chronic disease on a global scale and increase resistance to viruses like SARS-CoV-2.
Collapse
Affiliation(s)
- Daan L. de Frel
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe E. Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Douwe E. Atsma
| | - Hanno Pijl
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
| | - Jacob C. Seidell
- Department of Health Sciences, VU Medical Center, Amsterdam, Netherlands
| | - Pieter J. M. Leenen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Clinical Immunology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Elisabeth F. C. van Rossum
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
111
|
Zynat J, Li S, Ma Y, Han L, Ma F, Zhang Y, Xing B, Wang X, Guo Y. Impact of Abdominal Obesity on Thyroid Auto-Antibody Positivity: Abdominal Obesity Can Enhance the Risk of Thyroid Autoimmunity in Men. Int J Endocrinol 2020; 2020:6816198. [PMID: 32256575 PMCID: PMC7093900 DOI: 10.1155/2020/6816198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/29/2019] [Accepted: 12/06/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The interrelation between obesity and autoimmune thyroid diseases is complex and has not been confirmed. The aim of the present study was to observe the relationship between thyroid autoimmunity and obesity, especially abdominal obesity, in a large population. METHODS A total of 2253 residents who had lived in Xinjiang for more than 3 years were enrolled. Serum thyroid hormone concentration, thyroid autoantibodies, lipid parameters, Weight, height, and waist and hip circumference were measured. RESULTS The prevalence of thyroid peroxidase antibody (TPOAb) and/or thyroglobulin antibody (TgAb) positive was 32.1% (21.2% in men and 37% in women, P < 0.01). Compared with women, men had significantly higher TG levels, waist circumference, and hip circumference levels (P < 0.01), while women showed higher TSH, TPOAb, and TgAb levels (P < 0.01). The prevalence of overweight and obesity was 71.1% in men and 63.5% in women. Men had a higher prevalence of abdominal obesity than women (56.6% in men and 47.6% in women, P < 0.01). TPOAb correlates positively with waist circumference (r = 0.100, P < 0.05) in men. Binary logistic analysis showed that TPOAb positivity had increased risks of abdominal obesity in men, and the OR was 1.1044 (95% CI 1.035, 1.151, P < 0.05). CONCLUSION Our results indicate that men had higher lipid levels, thicker waist circumference, and higher prevalence of overweight, obesity, and abdominal obesity. Abdominal obesity is a risk factor for TPOAb positivity in men, suggesting that abdominal obesity can enhance the risk of thyroid autoimmunity in men.
Collapse
Affiliation(s)
- Jazyra Zynat
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Suli Li
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Yanrong Ma
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Li Han
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Fuhui Ma
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Yuyuan Zhang
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Bei Xing
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Xinling Wang
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| | - Yanying Guo
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang, China
| |
Collapse
|
112
|
Ducloux D, Courivaud C, Bamoulid J, Crepin T, Gaiffe E, Laheurte C, Vauchy C, Rebibou JM, Saas P, Borot S. Immune phenotype predicts new onset diabetes after kidney transplantation. Hum Immunol 2019; 80:937-942. [DOI: 10.1016/j.humimm.2019.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022]
|
113
|
Li H, Wetchapinant C, Zhang L, Wu K. High-Fat Diet from Weaning until Early Adulthood Impairs T Cell Development in the Thymus. Lipids 2019; 55:35-44. [PMID: 31608450 DOI: 10.1002/lipd.12193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/27/2022]
Abstract
The growing prevalence of childhood obesity has become a serious health problem over the past few decades. Although the immune system is greatly affected by childhood obesity, whether obesity influences the T cell development in the thymus is poorly understood. In this study, we used a high-fat diet (HFD)-induced obesity mice model to study the influence of HFD from weaning on the thymus. C57BL/6 mice (male, 3 weeks old) were fed a HFD or standard diet (lean controls) for 6 weeks. The bodyweight of mice fed with an HFD was 28% higher than that in the control group, while the thymus weight of HFD mice decreased by 15% compared with controls. As expected, thymic triacylglycerol content of the HFD mice increased by 37% compared to the control mice. Importantly, the frequencies of CD4+ and CD8+ single-positive (SP) thymocytes decreased by 38% and 44%, respectively. The apoptotic analysis revealed that thymic tissue of HFD mice had a higher level of Annexin-V positive thymocytes than control animals. Furthermore, the immunoblotting analysis showed that survival signal pathways in the thymus were impaired in the HFD mice, including the AKT/mTOR and ERK pathways. With the analysis of T-cell receptor excision circles (TREC), we found that HFD-induced obesity decreased recent thymic emigrants in spleen tissue. Our findings indicate that HFD from the weaning period impairs T cell development in the thymus, possibly by induction of apoptosis of thymocytes, involving disruption of survival signal pathways.
Collapse
Affiliation(s)
- Hao Li
- Department of Pathophysiology, Navy Medical University, 800 Xiangyin road, Shanghai, 200433, China
| | | | - Liya Zhang
- Baodi Clinical College of Tianjin Medical University, 8 Guangchuan road, Tianjin, 301800, China
| | - Kunpeng Wu
- Fudan University, 220 Handan road, Shanghai, 200433, China.,Department of Hematology, Huashan Hospital, Fudan University, 12 middle urumqi road, Shanghai, 200040, China
| |
Collapse
|
114
|
Zhang C, Zhang J, Liu W, Chen X, Liu Z, Zhou Z. Improvements in humoral immune function and glucolipid metabolism after laparoscopic sleeve gastrectomy in patients with obesity. Surg Obes Relat Dis 2019; 15:1455-1463. [DOI: 10.1016/j.soard.2019.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 02/08/2023]
|
115
|
Abstract
The contributions of the peripheral adaptive and innate immune systems to CNS autoimmunity have been extensively studied. However, the role of thymic selection in these conditions is much less well understood. The thymus is the primary lymphoid organ for the generation of T cells; thymic mechanisms ensure that cells with an overt autoreactive specificity are eliminated before they emigrate to the periphery and control the generation of thymic regulatory T cells. Evidence from animal studies demonstrates that thymic T cell selection is important for establishing tolerance to autoantigens. However, there is a considerable knowledge gap regarding the role of thymic selection in autoimmune conditions of the human CNS. In this Review, we critically examine the current body of experimental evidence for the contribution of thymic tolerance to CNS autoimmune diseases. An understanding of why dysfunction of either thymic or peripheral tolerance mechanisms rarely leads to CNS inflammation is currently lacking. We examine the potential of de novo T cell formation and thymic selection as novel therapeutic avenues and highlight areas for future study that are likely to make these targets the focus of future treatments.
Collapse
|
116
|
Abstract
Obesity is one of the leading causes of preventable mortalities in many parts of the globe. The rise in geriatric population due to better treatment opportunities has also emerged as a major public health challenge. Both of these health challenges have impacted developed as well as developing countries. Obesity is attributed as a powerful risk factor of a variety of health problems such as cardiovascular diseases, hypertension, type 2 diabetes, dementia, neuropsychiatric diseases and many more. On the other hand, ageing is a natural process involving a gradual decline in physiological functions and is associated with similar co-morbidities as obesity. This review discusses about the commonalities (termed as ‘Obesageing') between the pathological phenomenon of obesity and normal physiological process of ageing. A unique rodent model of obesageing has been developed (WNIN/Ob) that has characteristics of morbid obesity as well as premature ageing. Such a novel animal model would facilitate the understanding of the complex interplay of different mechanisms that are common to obesity and ageing and help to devise strategies in future to tackle the growing burden of obesity and ageing.
Collapse
Affiliation(s)
- Shampa Ghosh
- Endocrinology and Metabolism Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Jitendra Kumar Sinha
- Endocrinology and Metabolism Division, ICMR-National Institute of Nutrition, Hyderabad; Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Manchala Raghunath
- Endocrinology and Metabolism Division, ICMR-National Institute of Nutrition, Hyderabad, India
| |
Collapse
|
117
|
Naik A, Monjazeb AM, Decock J. The Obesity Paradox in Cancer, Tumor Immunology, and Immunotherapy: Potential Therapeutic Implications in Triple Negative Breast Cancer. Front Immunol 2019; 10:1940. [PMID: 31475003 PMCID: PMC6703078 DOI: 10.3389/fimmu.2019.01940] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/31/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer immunotherapy has been heralded as a breakthrough cancer treatment demonstrating tremendous success in improving tumor responses and survival of patients with hematological cancers and solid tumors. This novel promising treatment approach has in particular triggered optimism for triple negative breast cancer (TNBC) treatment, a subtype of breast cancer with distinct clinical features and poor clinical outcome. In early 2019, the FDA granted the first approval of immune checkpoint therapy, targeting PD-L1 (Atezolizumab) in combination with chemotherapy for the treatment of patients with locally advanced or metastatic PD-L1 positive TNBC. The efficacy of immuno-based interventions varies across cancer types and patient cohorts, which is attributed to a variety of lifestyle, clinical, and pathological factors. For instance, obesity has emerged as a risk factor for a dampened anti-tumor immune response and increased risk of immunotherapy-induced immune-related adverse events (irAEs) but has also been linked to improved outcomes with checkpoint blockade. Given the breadth of the rising global obesity epidemic, it is imperative to gain insight into the immunomodulatory effects of obesity in the peripheral circulation and within the tumor microenvironment. In this review, we resolve the impact of obesity on breast tumorigenesis and progression on the one hand, and on the immune contexture on the other hand. Finally, we speculate on the potential implications of obesity on immunotherapy response in breast cancer. This review clearly highlights the need for in vivo obese cancer models and representative clinical cohorts for evaluation of immunotherapy efficacy.
Collapse
Affiliation(s)
- Adviti Naik
- Qatar Foundation (QF), Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Arta Monir Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, University of California, Sacramento, Sacramento, CA, United States
| | - Julie Decock
- Qatar Foundation (QF), Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
118
|
Crouch MJ, Kosaraju R, Guesdon W, Armstrong M, Reisdorph N, Jain R, Fenton J, Shaikh SR. Frontline Science: A reduction in DHA-derived mediators in male obesity contributes toward defects in select B cell subsets and circulating antibody. J Leukoc Biol 2019; 106:241-257. [PMID: 30576001 PMCID: PMC10020993 DOI: 10.1002/jlb.3hi1017-405rr] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/20/2018] [Accepted: 12/11/2018] [Indexed: 01/25/2023] Open
Abstract
Obesity dysregulates B cell populations, which contributes toward poor immunological outcomes. We previously reported that differing B cell subsets are lowered in the bone marrow of obese male mice. Here, we focused on how lipid metabolites synthesized from docosahexaenoic acid (DHA) known as specialized pro-resolving lipid mediators (SPMs) influence specific B cell populations in obese male mice. Metabololipidomics revealed that splenic SPM precursors 14-hydroxydocosahexaenoic acid (14-HDHA), 17-hydroxydocosahexaenoic acid (17-HDHA), and downstream protectin DX (PDX) were decreased in obese male C57BL/6J mice. Simultaneous administration of these mediators to obese mice rescued major decrements in bone marrow B cells, modest impairments in the spleen, and circulating IgG2c, which is pro-inflammatory in obesity. In vitro studies with B cells, flow cytometry experiments with ALOX5-/- mice, and lipidomic analyses revealed the lowering of 14-HDHA/17-HDHA/PDX and dysregulation of B cell populations in obesity was driven indirectly via B cell extrinsic mechanisms. Notably, the lowering of lipid mediators was associated with an increase in the abundance of n-6 polyunsaturated fatty acids, which have a high affinity for SPM-generating enzymes. Subsequent experiments revealed female obese mice generally maintained the levels of SPM precursors, B cell subsets, and antibody levels. Finally, obese human females had increased circulating plasma cells accompanied by ex vivo B cell TNFα and IL-10 secretion. Collectively, the data demonstrate that DHA-derived mediators of the SPM pathway control the number of B cell subsets and pro-inflammatory antibody levels in obese male but not female mice through a defect that is extrinsic to B cells.
Collapse
Affiliation(s)
- Miranda J Crouch
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rasagna Kosaraju
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - William Guesdon
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado, Denver, Colorado, USA
| | - Raghav Jain
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Jenifer Fenton
- The College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
119
|
Li G, Petkova TD, Laritsky E, Kessler N, Baker MS, Zhu S, Waterland RA. Early postnatal overnutrition accelerates aging-associated epigenetic drift in pancreatic islets. ENVIRONMENTAL EPIGENETICS 2019; 5:dvz015. [PMID: 31528363 PMCID: PMC6735752 DOI: 10.1093/eep/dvz015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 05/02/2023]
Abstract
Pancreatic islets of type 2 diabetes patients have altered DNA methylation, contributing to islet dysfunction and the onset of type 2 diabetes. The cause of these epigenetic alterations is largely unknown. We set out to test whether (i) islet DNA methylation would change with aging and (ii) early postnatal overnutrition would persistently alter DNA methylation. We performed genome-scale DNA methylation profiling in islets from postnatally over-nourished (suckled in a small litter) and control male mice at both postnatal day 21 and postnatal day 180. DNA methylation differences were validated using quantitative bisulfite pyrosequencing, and associations with expression were assessed by RT-PCR. We discovered that genomic regions that are hypermethylated in exocrine relative to endocrine pancreas tend to gain methylation in islets during aging (R 2 = 0.33, P < 0.0001). These methylation differences were inversely correlated with mRNA expression of genes relevant to β cell function [including Rab3b (Ras-related protein Rab-3B), Cacnb3 (voltage-dependent L-type calcium channel subunit 3), Atp2a3 (sarcoplasmic/endoplasmic reticulum calcium ATPase 3) and Ins2 (insulin 2)]. Relative to control, small litter islets showed DNA methylation differences directly after weaning and in adulthood, but few of these were present at both ages. Surprisingly, we found substantial overlap of methylated loci caused by aging and small litter feeding, suggesting that the age-associated gain of DNA methylation happened much earlier in small litter islets than control islets. Our results provide the novel insights that aging-associated DNA methylation increases reflect an epigenetic drift toward the exocrine pancreas epigenome, and that early postnatal overnutrition may accelerate this process.
Collapse
Affiliation(s)
- Ge Li
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Tihomira D Petkova
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Eleonora Laritsky
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Noah Kessler
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Maria S Baker
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Shaoyu Zhu
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
| | - Robert A Waterland
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, Houston, TX, USA
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Correspondence address. Departments of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, USDA/ARS Children’s Nutrition Research Center, 1100 Bates Street, Ste. 5080, Houston, TX 77030, USA. Tel: +1-713-798-0304; E-mail:
| |
Collapse
|
120
|
Mahtab S, Kar P, Saha S, Sreenivas V, Sottini A, Imberti L, Goswami R. Central Immune Tolerance of T and B Cells in Patients With Idiopathic Hypoparathyroidism, T1D, and Autoimmune Thyroiditis. J Endocr Soc 2019; 3:1175-1184. [PMID: 31139764 PMCID: PMC6532674 DOI: 10.1210/js.2018-00344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/15/2019] [Indexed: 02/07/2023] Open
Abstract
CONTEXT Pathogenesis of idiopathic hypoparathyroidism (IH) is under investigation. Abnormalities in central immune tolerance have yet not been investigated in this condition. T-cell receptor excision circles (TRECs) and kappa-deleting recombination excision circles (KRECs), formed during receptor gene rearrangements, are tools to assess central T- and B-cell output. OBJECTIVE We assessed the number of circulating TRECs and KRECs in patients with IH, autoimmune type 1 diabetes (T1D), and autoimmune thyroiditis (ATs) and healthy controls (HCs). DESIGN Comparative case-control at tertiary care center. SUBJECTS AND METHODS Absolute and relative TRECs and KRECs were measured in DNA extracted from whole blood of patients with IH (n = 181, 22 of whom were reassessed after a decade of follow-up) and T1D (n = 133), AT (n = 53), and HC (n = 135) using a quantitative real-time PCR/TaqMan® probe technique. RESULTS Absolute and relative means of TRECs and KRECs in IH were comparable to HCs, and no differences were found between IH with and without calcium-sensing receptor antibodies or class I HLA-A*26:01 association. TRECs and KRECs did not change after a decade of follow-up. T1D had significantly higher absolute TRECs than IH, AT, and HCs, whereas AT patients showed lower TRECs and the highest KRECs; these levels showed no noteworthy correlation with thyroid dysfunctions. CONCLUSION Patients with IH showed TRECs and KRECs comparable to HCs, indicating an intact mechanism of T- and B-cell central immune tolerance. Interestingly, absolute TRECs were significantly higher in T1D than HCs, suggesting impaired central immune tolerance in T1D.
Collapse
Affiliation(s)
- Samrina Mahtab
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Parmita Kar
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Soma Saha
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | | | - Alessandra Sottini
- Diagnostic Department, Centro di Ricerca Emato-oncologica AIL, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Luisa Imberti
- Diagnostic Department, Centro di Ricerca Emato-oncologica AIL, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Ravinder Goswami
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
121
|
Chang MR, Ciesla A, Strutzenberg TS, Novick SJ, He Y, Garcia-Ordonez RD, Frkic RL, Bruning JB, Kamenecka TM, Griffin PR. Unique Polypharmacology Nuclear Receptor Modulator Blocks Inflammatory Signaling Pathways. ACS Chem Biol 2019; 14:1051-1062. [PMID: 30951276 DOI: 10.1021/acschembio.9b00236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity and rheumatic disease are mechanistically linked via chronic inflammation. The orphan receptor TREM-1 (triggering receptor expressed on myeloid cells-1) is a potent amplifier of proinflammatory and noninfectious immune responses. Here, we show that the pan modulator SR1903 effectively blocks TREM-1 activation. SR1903 emerged from a chemical series of potent RORγ inverse agonists, although unlike close structural analogues, it has modest agonist activity on LXR and weak repressive activity (inverse agonism) of PPARγ, three receptors that play essential roles in inflammation and metabolism. The anti-inflammatory and antidiabetic efficacy of this unique modulator in collagen-induced arthritis and diet-induced obesity mouse models is demonstrated. Interestingly, in the context of obesity, SR1903 aided in the maintenance of the thymic homeostasis unlike selective RORγ inverse agonists. SR1903 was well-tolerated following chronic administration, and combined, these data suggest that it may represent a viable strategy for treatment of both metabolic and inflammatory disease. More importantly, the ability of SR1903 to block LPS signaling suggests the potential utility of this unique polypharmacological modulator for treatment of innate immune response disorders.
Collapse
Affiliation(s)
- Mi Ra Chang
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Anthony Ciesla
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Timothy S. Strutzenberg
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Scott J. Novick
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Yuanjun He
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Ruben D. Garcia-Ordonez
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Rebecca L. Frkic
- Institute for Photonics & Advanced Sensing (IPAS), School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - John B. Bruning
- Institute for Photonics & Advanced Sensing (IPAS), School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Theodore M. Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Patrick R. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida 33458, United States
| |
Collapse
|
122
|
Honce R, Schultz-Cherry S. Impact of Obesity on Influenza A Virus Pathogenesis, Immune Response, and Evolution. Front Immunol 2019; 10:1071. [PMID: 31134099 PMCID: PMC6523028 DOI: 10.3389/fimmu.2019.01071] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
With the rising prevalence of obesity has come an increasing awareness of its impact on communicable disease. As a consequence of the 2009 H1N1 influenza A virus pandemic, obesity was identified for the first time as a risk factor for increased disease severity and mortality in infected individuals. Over-nutrition that results in obesity causes a chronic state of meta-inflammation with systemic implications for immunity. Obese hosts exhibit delayed and blunted antiviral responses to influenza virus infection, and they experience poor recovery from the disease. Furthermore, the efficacy of antivirals and vaccines is reduced in this population and obesity may also play a role in altering the viral life cycle, thus complementing the already weakened immune response and leading to severe pathogenesis. Case studies and basic research in human cohorts and animal models have highlighted the prolonged viral shed in the obese host, as well as a microenvironment that permits the emergence of virulent minor variants. This review focuses on influenza A virus pathogenesis in the obese host, and on the impact of obesity on the antiviral response, viral shed, and viral evolution. We comprehensively analyze the recent literature on how and why viral pathogenesis is altered in the obese host along with the impact of the altered host and pathogenic state on viral evolutionary dynamics in multiple models. Finally, we summarized the effectiveness of current vaccines and antivirals in this populations and the questions that remain to be answered. If current trends continue, nearly 50% of the worldwide population is projected to be obese by 2050. This population will have a growing impact on both non-communicable and communicable diseases and may affect global evolutionary trends of influenza virus.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, United States
- Integrated Program in Biomedical Sciences, Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
123
|
Honce R, Schultz-Cherry S. Influenza in obese travellers: increased risk and complications, decreased vaccine effectiveness. J Travel Med 2019; 26:taz020. [PMID: 30924873 PMCID: PMC6509472 DOI: 10.1093/jtm/taz020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Obesity is a worldwide epidemic and was empirically shown to increase the risk of developing severe influenza virus infection. As international travel becomes more common and obesity is now prevalent even in low- and middle-income countries, travellers may have an increased risk of contracting influenza virus especially during peak influenza season. METHODS An analysis of the literature, centred on publications from 2014-19, was performed, with an emphasis on human epidemiological data, human studies ex vivo and studies in mouse models of obesity. Our search efforts focused on influenza disease severity, pathogenesis, evolutionary dynamics and measures of infection control in the obese and overweight host. RESULTS Obesity is associated with an increased risk of infection, as well as a greater chance for hospitalization and severe complications. Studies in mouse models of obesity have uncovered that obese hosts suffer increased viral spread, delayed viral clearance and heightened damage to the respiratory epithelium. Innate and adaptive immune responses are delayed, thus increasing morbidity and mortality. Further, infection control measures, including vaccination and antivirals, prove less effective in obese hosts. Finally, the obese microenvironment allows for increased duration and amount of viral shedding and potentially increases the chance for emergence of virulent minor variants in the viral population. Together, obese hosts are at high risk of influenza infection, as well as severe sequelae following infection. CONCLUSION Obese travellers should be aware of influenza activity in the regions visited, as well as take protective measures prior to travel. Vaccination is highly recommended for all travellers, but especially highly susceptible obese travellers.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
124
|
|
125
|
Gender Dictates the Relationship between Serum Lipids and Leukocyte Counts in the National Health and Nutrition Examination Survey 1999⁻2004. J Clin Med 2019; 8:jcm8030365. [PMID: 30875952 PMCID: PMC6463027 DOI: 10.3390/jcm8030365] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/10/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
Dyslipidemias and leukocytosis are associated with cardiovascular disease and immune disorders. Mechanistic studies have shown lipoprotein metabolism to play a significant role in the regulation of atherosclerosis development and leukocyte activation, whereas lipid-lowering treatments have been shown to exert beneficial anti-inflammatory and immunomodulatory effects in clinical trials. However, the relationship between clinical markers of lipid metabolism and leukocyte counts has not been extensively evaluated at the population level. We aimed to determine whether clinical blood lipid measures are associated with leukocyte counts in the general U.S. population represented in the National Health and Nutrition Examination Survey (NHANES) 1999–2004, and whether differences exist between men and women (n = 5647). We observed a strong positive linear trend between serum triglycerides vs. blood lymphocyte and basophil counts in both men and women, whereas a positive trend between monocytes vs. triglycerides and lymphocytes vs. total cholesterol and LDL-cholesterol (LDL-C) was only detected in women. Conversely, HDL-C was inversely associated with a greater number of leukocyte subsets in men, whereas inverse trends between HDL-C vs. lymphocytes were observed in both men and women. In multiple regression models, a 10% increase in total cholesterol, LDL-C, and triglycerides was associated with a predicted 1.6%, 0.6%, and 1.4% increase in blood lymphocyte counts in women, respectively, whereas no relationship was observed in men. In both men and women, a 10% increase in triglycerides was additionally associated with higher lymphocyte, neutrophil, and basophil counts, whereas 10% increases in HDL-cholesterol were associated with significantly lower lymphocyte, neutrophil, eosinophil, and basophil counts in men, in addition to lower lymphocyte and monocyte counts in women. These findings suggest that clinical lipid markers may be used to predict blood leukocyte distributions, and that a gender-specific relationship exists between distinct classes of serum lipids and immune cell subsets.
Collapse
|
126
|
Abstract
Obesity is associated with both increased cancer incidence and progression in multiple tumour types, and is estimated to contribute to up to 20% of cancer-related deaths. These associations are driven, in part, by metabolic and inflammatory changes in adipose tissue that disrupt physiological homeostasis both within local tissues and systemically. However, the mechanisms underlying the obesity-cancer relationship are poorly understood. In this Review, we describe how the adipose tissue microenvironment (ATME) evolves during body-weight gain, and how these changes might influence tumour initiation and progression. We focus on multiple facets of ATME physiology, including inflammation, vascularity and fibrosis, and discuss therapeutic interventions that have the potential to normalize the ATME, which might be translationally relevant for cancer prevention and therapy. Given that the prevalence of obesity is increasing on an international scale, translational research initiatives are urgently needed to provide mechanistic explanations for the obesity-cancer relationship, and how to best identify high-risk individuals without relying on crude measures, such as BMI.
Collapse
Affiliation(s)
- Daniela F Quail
- Goodman Cancer Research Centre, Department of Physiology, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
127
|
Banfai K, Ernszt D, Pap A, Bai P, Garai K, Belharazem D, Pongracz JE, Kvell K. "Beige" Cross Talk Between the Immune System and Metabolism. Front Endocrinol (Lausanne) 2019; 10:369. [PMID: 31275241 PMCID: PMC6591453 DOI: 10.3389/fendo.2019.00369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/24/2019] [Indexed: 12/25/2022] Open
Abstract
With thymic senescence the epithelial network shrinks to be replaced by adipose tissue. Transcription factor TBX-1 controls thymus organogenesis, however, the same TBX-1 has also been reported to orchestrate beige adipose tissue development. Given these different roles of TBX-1, we have assessed if thymic TBX-1 expression persists and demonstrates this dualism during adulthood. We have also checked whether thymic adipose involution could yield beige adipose tissue. We have used adult mouse and human thymus tissue from various ages to evaluate the kinetics of TBX-1 expression, as well as mouse (TEP1) and human (1889c) thymic epithelial cells (TECs) for our studies. Electron micrographs show multi-locular lipid deposits typical of beige adipose cells. Histology staining shows the accumulation of neutral lipid deposits. qPCR measurements show persistent and/or elevating levels of beige-specific and beige-indicative markers (TBX-1, EAR-2, UCP-1, PPAR-gamma). We have performed miRNome profiling using qPCR-based QuantStudio platform and amplification-free NanoString platform. We have observed characteristic alterations, including increased miR21 level (promoting adipose tissue development) and decreased miR34a level (bias toward beige adipose tissue differentiation). Finally, using the Seahorse metabolic platform we have recorded a metabolic profile (OCR/ECAR ratio) indicative of beige adipose tissue. In summary, our results support that thymic adipose tissue emerging with senescence is bona fide beige adipose tissue. Our data show how the borders blur between a key immune tissue (the thymus) and a key metabolic tissue (beige adipose tissue) with senescence. Our work contributes to the understanding of cross talk between the immune system and metabolism.
Collapse
Affiliation(s)
- Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - David Ernszt
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- Department of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Bai
- Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
- MTA-DE Lendulet Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Djeda Belharazem
- Department of Pathology, University Hospital of Mannheim, Mannheim, Germany
| | - Judit E. Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- *Correspondence: Krisztian Kvell
| |
Collapse
|
128
|
McDonnell WJ, Koethe JR, Mallal SA, Pilkinton MA, Kirabo A, Ameka MK, Cottam MA, Hasty AH, Kennedy AJ. High CD8 T-Cell Receptor Clonality and Altered CDR3 Properties Are Associated With Elevated Isolevuglandins in Adipose Tissue During Diet-Induced Obesity. Diabetes 2018; 67:2361-2376. [PMID: 30181158 PMCID: PMC6198339 DOI: 10.2337/db18-0040] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/20/2018] [Indexed: 12/29/2022]
Abstract
Adipose tissue (AT) CD4+ and CD8+ T cells contribute to obesity-associated insulin resistance. Prior studies identified conserved T-cell receptor (TCR) chain families in obese AT, but the presence and clonal expansion of specific TCR sequences in obesity has not been assessed. We characterized AT and liver CD8+ and CD4+ TCR repertoires of mice fed a low-fat diet (LFD) and high-fat diet (HFD) using deep sequencing of the TCRβ chain to quantify clonal expansion, gene usage, and CDR3 sequence. In AT CD8+ T cells, HFD reduced TCR diversity, increased the prevalence of public TCR clonotypes, and selected for TCR CDR3 regions enriched in positively charged and less polarized amino acids. Although TCR repertoire alone could distinguish between LFD- and HFD-fed mice, these properties of the CDR3 region of AT CD8+ T cells from HFD-fed mice led us to examine the role of negatively charged and nonpolar isolevuglandin (isoLG) adduct-containing antigen-presenting cells within AT. IsoLG-adducted protein species were significantly higher in AT macrophages of HFD-fed mice; isoLGs were elevated in M2-polarized macrophages, promoting CD8+ T-cell activation. Our findings demonstrate that clonal TCR expansion that favors positively charged CDR3s accompanies HFD-induced obesity, which may be an antigen-driven response to isoLG accumulation in macrophages.
Collapse
Affiliation(s)
- Wyatt J McDonnell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN
| | - John R Koethe
- Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN
| | - Simon A Mallal
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Mark A Pilkinton
- Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Magdalene K Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alyssa H Hasty
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Arion J Kennedy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
129
|
Apelin/APJ system: A novel promising target for anti-aging intervention. Clin Chim Acta 2018; 487:233-240. [PMID: 30296443 DOI: 10.1016/j.cca.2018.10.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/04/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Apelin, an endogenous ligand for the G protein-coupled receptor APJ, is widely expressed in various organs. Recent research has indicated that the Apelin/APJ system plays an important role in aging. Apelin and APJ receptor expression are down-regulated with increasing age. In murine models, Apelin and APJ knockouts exhibit accelerated senescence whereas Apelin-restoration results in enhanced vigor and rejuvenated behavioral and circadian phenotypes. Furthermore, aged Apelin knockout mice develop progressive impairment of cardiac contractility associated with systolic dysfunction. Apelin is crucial to maintain cardiac contractility in aging. Moreover, the Apelin/APJ system appears to be involved in regulation of renin-angiotensin-aldosterone system (RAAS), apoptosis, inflammation and oxidative stress which promotes aging. Likewise, the Apelin/APJ system regulates autophagy, stem cells and the sirtuin family thus contributing to anti-aging. In this review, we describe the relationship between Apelin/APJ system and aging. We elaborate on the role of the Apelin/APJ system in aging stimulators, aging inhibitors and age-related diseases such as obesity, diabetes and cardiovascular disease. We conclude that Apelin/APJ system might become a novel promising therapeutic target for anti-aging.
Collapse
|
130
|
Clave E, Araujo IL, Alanio C, Patin E, Bergstedt J, Urrutia A, Lopez-Lastra S, Li Y, Charbit B, MacPherson CR, Hasan M, Melo-Lima BL, Douay C, Saut N, Germain M, Trégouët DA, Morange PE, Fontes M, Duffy D, Di Santo JP, Quintana-Murci L, Albert ML, Toubert A. Human thymopoiesis is influenced by a common genetic variant within the TCRA-TCRD locus. Sci Transl Med 2018; 10:10/457/eaao2966. [DOI: 10.1126/scitranslmed.aao2966] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 04/11/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The thymus is the primary lymphoid organ where naïve T cells are generated; however, with the exception of age, the parameters that govern its function in healthy humans remain unknown. We characterized the variability of thymic function among 1000 age- and sex-stratified healthy adults of the Milieu Intérieur cohort, using quantification of T cell receptor excision circles (TRECs) in peripheral blood T cells as a surrogate marker of thymopoiesis. Age and sex were the only nonheritable factors identified that affect thymic function. TREC amounts decreased with age and were higher in women compared to men. In addition, a genome-wide association study revealed a common variant (rs2204985) within the T cell receptor TCRA-TCRD locus, between the DD2 and DD3 gene segments, which associated with TREC amounts. Strikingly, transplantation of human hematopoietic stem cells with the rs2204985 GG genotype into immunodeficient mice led to thymopoiesis with higher TRECs, increased thymocyte counts, and a higher TCR repertoire diversity. Our population immunology approach revealed a genetic locus that influences thymopoiesis in healthy adults, with potentially broad implications in precision medicine.
Collapse
|
131
|
Canter RJ, Le CT, Beerthuijzen JM, Murphy WJ. Obesity as an immune-modifying factor in cancer immunotherapy. J Leukoc Biol 2018; 104:487-497. [PMID: 29762866 PMCID: PMC6113103 DOI: 10.1002/jlb.5ri1017-401rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy has achieved breakthrough status in many advanced stage malignancies and is rapidly becoming the fourth arm of cancer treatment. Although cancer immunotherapy has generated significant excitement because of the potential for complete and sometimes durable responses, there is also the potential for severe and occasionally life-threatening toxicities, including cytokine release syndrome and severe autoimmunity. A large body of work also points to a "metainflammatory" state in obesity associated with impairment of immune responses. Because immune checkpoint blockade (and other cancer immunotherapies) have altered the landscape of immunotherapy in cancer, it is important to understand how immune responses are shaped by obesity and how obesity may modify both immunotherapy responses and potential toxicities.
Collapse
Affiliation(s)
- Robert J. Canter
- University of California, Davis, School of Medicine, Department of Surgery, Division of Surgical Oncology, Sacramento, CA 95817
| | - Catherine T Le
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| | - Johanna M.T. Beerthuijzen
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| | - William J. Murphy
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| |
Collapse
|
132
|
Diet-induced obese mice exhibit altered immune responses to early Salmonella Typhimurium oral infection. J Microbiol 2018; 56:673-682. [PMID: 30141160 DOI: 10.1007/s12275-018-8083-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 10/28/2022]
Abstract
Obesity is a chronic disease associated with different metabolic diseases as well as alterations in immune cell function. It is characterized by a chronic systemic low grade inflammation. There are several studies demonstrating the influence of obesity on the impaired immune response to infection. However, it is not completely clear whether the obese environment influences the development or maintenance of the immune response against infections. The aim of this study was to determine how obesity induced by a high-fat diet affects the immune response to an early oral Salmonella infection. Four groups of mice were kept in separate cages. Two of these designated as controls, fed with a normal diet; whereas other two groups were fed with a high fat diet for 10 weeks. Some mice were used for Salmonella oral infection. After 7 days of oral infection with S. Thypimurium the proportions of spleen cell subsets expressing activation markers in normal diet and HFD obese mice were stained with monoclonal antibodies and analyzed by flow cytometry. Also, mRNA levels of different cytokines were quantified by RT-PCR. It was found that obesity affects the function of the immune system against an early oral Salmonella infection, decreasing NK cells, altering the expression of activation molecules as well as cytokines mRNA levels. Interestingly, the expression some activation molecules on T lymphocytes was reestablished after Salmonella infection, but not the CD25 expression. Immune alterations could lead to immunosuppression or increased susceptibility to infections in HFD obese mice.
Collapse
|
133
|
Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 2018; 19:447-459. [PMID: 30054761 PMCID: PMC6223730 DOI: 10.1007/s10522-018-9763-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/21/2018] [Indexed: 12/13/2022]
Abstract
Cellular senescence is now considered as a major mechanism in the development and progression of various diseases and this may include metabolic diseases such as obesity and type-2 diabetes. The presence of obesity and diabetes is a major risk factor in the development of additional health conditions, such as cardiovascular disease, kidney disease and cancer. Since senescent cells can drive disease development, obesity and diabetes can potentially create an environment that accelerates cell senescence within other tissues of the body. This can consequently manifest as age-related biological impairments and secondary diseases. Cell senescence in cell types linked with obesity and diabetes, namely adipocytes and pancreatic beta cells will be explored, followed by a discussion on the role of obesity and diabetes in accelerating ageing through induction of premature cell senescence mediated by high glucose levels and oxidised low-density lipoproteins. Particular emphasis will be placed on accelerated cell senescence in endothelial progenitor cells, endothelial cells and vascular smooth muscle cells with relation to cardiovascular disease and proximal tubular cells with relation to kidney disease. A summary of the potential strategies for therapeutically targeting senescent cells for improving health is also presented.
Collapse
|
134
|
Mu WC, VanHoosier E, Elks CM, Grant RW. Long-Term Effects of Dietary Protein and Branched-Chain Amino Acids on Metabolism and Inflammation in Mice. Nutrients 2018; 10:nu10070918. [PMID: 30021962 PMCID: PMC6073443 DOI: 10.3390/nu10070918] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Aging is the main factor involved in the onset of degenerative diseases. Dietary protein restriction has been shown to increase the lifespan of rodents and improve metabolic phenotype. Branched-chain amino acids (BCAA) can act as nutrient signals that increase the lifespan of mice after prolonged supplementation. It remains unclear whether the combination of protein restriction and BCAA supplementation improves metabolic and immunological profiles during aging. Here, we investigated how dietary protein levels and BCAA supplementation impact metabolism and immune profile during a 12-month intervention in adult male C57BL/6J mice. We found that protein restriction improved insulin tolerance and increased hepatic fibroblast growth factor 21 mRNA, circulating interleukin (IL)-5 concentration, and thermogenic uncoupling protein 1 in subcutaneous white fat. Surprisingly, BCAA supplementation conditionally increased body weight, lean mass, and fat mass, and deteriorated insulin intolerance during protein restriction, but not during protein sufficiency. BCAA also induced pro-inflammatory gene expression in visceral adipose tissue under both normal and low protein conditions. These results suggest that dietary protein levels and BCAA supplementation coordinate a complex regulation of metabolism and tissue inflammation during prolonged feeding.
Collapse
MESH Headings
- Adiposity
- Aging
- Amino Acids, Branched-Chain/adverse effects
- Amino Acids, Branched-Chain/metabolism
- Amino Acids, Branched-Chain/therapeutic use
- Animals
- Cytokines/blood
- Diet, Protein-Restricted/adverse effects
- Dietary Proteins/adverse effects
- Dietary Proteins/metabolism
- Dietary Proteins/therapeutic use
- Dietary Supplements/adverse effects
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Insulin Resistance
- Liver/growth & development
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Male
- Mice, Inbred C57BL
- Organ Size
- Proteomics/methods
- Random Allocation
- Sarcopenia/immunology
- Sarcopenia/metabolism
- Sarcopenia/pathology
- Sarcopenia/prevention & control
- Spleen/growth & development
- Spleen/immunology
- Spleen/metabolism
- Spleen/pathology
- Subcutaneous Fat, Abdominal/growth & development
- Subcutaneous Fat, Abdominal/immunology
- Subcutaneous Fat, Abdominal/metabolism
- Subcutaneous Fat, Abdominal/pathology
- Thymus Gland/growth & development
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Thymus Gland/pathology
- Weight Gain
Collapse
Affiliation(s)
- Wei-Chieh Mu
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Erin VanHoosier
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Carrie M Elks
- Matrix Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Ryan W Grant
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
135
|
Makki K, Deehan EC, Walter J, Bäckhed F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018; 23:705-715. [DOI: 10.1016/j.chom.2018.05.012] [Citation(s) in RCA: 949] [Impact Index Per Article: 158.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
136
|
Tencerova M, Figeac F, Ditzel N, Taipaleenmäki H, Nielsen TK, Kassem M. High-Fat Diet-Induced Obesity Promotes Expansion of Bone Marrow Adipose Tissue and Impairs Skeletal Stem Cell Functions in Mice. J Bone Miner Res 2018; 33:1154-1165. [PMID: 29444341 DOI: 10.1002/jbmr.3408] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022]
Abstract
Obesity represents a risk factor for development of insulin resistance and type 2 diabetes. In addition, it has been associated with increased adipocyte formation in the bone marrow (BM) along with increased risk for bone fragility fractures. However, little is known on the cellular mechanisms that link obesity, BM adiposity, and bone fragility. Thus, in an obesity intervention study in C57BL/6J mice fed with a high-fat diet (HFD) for 12 weeks, we investigated the molecular and cellular phenotype of bone marrow adipose tissue (BMAT), BM progenitor cells, and BM microenvironment in comparison to peripheral adipose tissue (AT). HFD decreased trabecular bone mass by 29%, cortical thickness by 5%, and increased BM adiposity by 184%. In contrast to peripheral AT, BMAT did not exhibit pro-inflammatory phenotype. BM progenitor cells isolated from HFD mice exhibited decreased mRNA levels of inflammatory genes (Tnfα, IL1β, Lcn2) and did not manifest an insulin resistant phenotype evidenced by normal levels of pAKT after insulin stimulation as well as normal levels of insulin signaling genes. In addition, BM progenitor cells manifested enhanced adipocyte differentiation in HFD condition. Thus, our data demonstrate that BMAT expansion in response to HFD exerts a deleterious effect on the skeleton. Continuous recruitment of progenitor cells to adipogenesis leads to progenitor cell exhaustion, decreased recruitment to osteoblastic cells, and decreased bone formation. In addition, the absence of insulin resistance and inflammation in the BM suggest that BMAT buffers extra energy in the form of triglycerides and thus plays a role in whole-body energy homeostasis. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense C, Denmark.,Danish Diabetes Academy, Odense C, Denmark
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense C, Denmark
| | - Nicholas Ditzel
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense C, Denmark
| | - Hanna Taipaleenmäki
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand, and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tina Kamilla Nielsen
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense C, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense C, Denmark.,Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen, Denmark.,Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Kingdom of Saudi Arabia
| |
Collapse
|
137
|
Padrón Á, Hurez V, Gupta HB, Clark CA, Pandeswara SL, Yuan B, Svatek RS, Turk MJ, Drerup JM, Li R, Curiel TJ. Age effects of distinct immune checkpoint blockade treatments in a mouse melanoma model. Exp Gerontol 2018; 105:146-154. [DOI: 10.1016/j.exger.2017.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/26/2017] [Accepted: 12/28/2017] [Indexed: 02/08/2023]
|
138
|
Ismail IA, El-Bakry HA, Soliman SS. Melatonin and tumeric ameliorate aging-induced changes: implication of immunoglobulins, cytokines, DJ-1/NRF2 and apoptosis regulation. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2018; 10:70-82. [PMID: 29755640 PMCID: PMC5943606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/05/2018] [Indexed: 06/08/2023]
Abstract
Aging is associated with several biological, physiological, cellular and histological changes. In the present study, we investigated the effect of aging on different signaling pathways, including antioxidant system, apoptosis and immune status. Several natural products were used to ameliorate and block aging-related changes. Melatonin and turmeric have been known to ameliorate and decrease aging-related changes. However, the exact mechanism(s) of their action is not fully understood. In the present study, we tried to uncover the regulatory mechanism(s) by which melatonin and turmeric work against aging. We found that aging differentially regulated blood serum immunoglobulins; increased IgA and decreased IgE. Furthermore, all the serum cytokines investigated (TNF-α, IFN-γ, IL-6 and IL-8) were highly increased by aging. In addition, the antioxidant upstream regulators; DJ-1 and NRF2 were markedly repressed with aging in thymus tissues. We also found that aging induced apoptosis promoting genes p53 and Bax mRNA in thymus tissues. Finally, we found clear histological changes in thymus and spleen tissues. Administration of either melatonin or tumeric clearly ameliorated and blocked to some extinct the effect of aging. Altogether, aging was associated with downregulation of antioxidant regulators; DJ-1 and NRF2, promoted apoptosis and induced changes in the immune status. Furthermore, melatonin and tumeric markedly reversed the action of aging through activating DJ-1/NRF2 signaling pathway and inhibiting p53/Bax apoptotic pathway.
Collapse
Affiliation(s)
- Ismail Ahmed Ismail
- Department of Biology, Faculty of Science, Taibah University, Yanbu BranchSaudi Arabia
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut UniversityAssiut 71516, Egypt
| | - Hanan A El-Bakry
- Department of Zoology, Faculty of Science, Minia UniversityEgypt
| | - Safaa S Soliman
- Department of Zoology, Faculty of Science, Minia UniversityEgypt
| |
Collapse
|
139
|
Aguilar EG, Murphy WJ. Obesity induced T cell dysfunction and implications for cancer immunotherapy. Curr Opin Immunol 2018; 51:181-186. [PMID: 29655021 DOI: 10.1016/j.coi.2018.03.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Obesity has been shown to increase risk for a number of different disorders, including cancer. In addition, obesity is also associated with immune dysfunction, which could contribute to its strong association with other comorbidities. Recently, the immune system has been found to be heavily regulated by changes in metabolism. In particular, T cells are able to respond to intrinsic metabolic regulatory mechanisms, as well as extrinsic factors such as the changes in metabolite availability. The dysfunctional metabolic environment created by obesity could therefore have a direct impact on T cell responses. In this review, we highlight recent findings in the fields of T cell biology and obesity, with a focus on mechanisms driving T cell dysfunction and potential implications for immunotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- Ethan G Aguilar
- Department of Dermatology, UC Davis School of Medicine, Sacramento, CA 95816, USA
| | - William J Murphy
- Department of Dermatology, UC Davis School of Medicine, Sacramento, CA 95816, USA; Department of Internal Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
140
|
Abstract
Obesity has been shown to increase risk for a number of different disorders, including cancer. In addition, obesity is also associated with immune dysfunction, which could contribute to its strong association with other comorbidities. Recently, the immune system has been found to be heavily regulated by changes in metabolism. In particular, T cells are able to respond to intrinsic metabolic regulatory mechanisms, as well as extrinsic factors such as the changes in metabolite availability. The dysfunctional metabolic environment created by obesity could therefore have a direct impact on T cell responses. In this review, we highlight recent findings in the fields of T cell biology and obesity, with a focus on mechanisms driving T cell dysfunction and potential implications for immunotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- Ethan G Aguilar
- Department of Dermatology, UC Davis School of Medicine, Sacramento, CA 95816, USA
| | - William J Murphy
- Department of Dermatology, UC Davis School of Medicine, Sacramento, CA 95816, USA; Department of Internal Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
141
|
Harrington KA, Kennedy DS, Tang B, Hickie C, Phelan E, Torreggiani W, Halpenny D. Computed tomographic evaluation of the thymus-does obesity affect thymic fatty involution in a healthy young adult population? Br J Radiol 2018; 91:20170609. [PMID: 29356558 DOI: 10.1259/bjr.20170609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To determine a relationship between increased body mass index (BMI) and fatty involution of the thymus in subjects aged between 20 and 30 years. METHODS CT images of 94 patients aged between 20 and 30 years were reviewed. Quantitative thymic mean attenuation was recorded and qualitative thymic attenuation was assigned to 1 of 4 possible grades. BMI and subcutaneous fat thickness were documented. Correlations between thymic attenuation, and BMI and subcutaneous fat thickness were assessed using linear regression models. Differences in thymic attenuation in overweight vs normal weight patients were assessed using t-test and Pearson Χ2 analysis. RESULTS Low mean thymic attenuation values were associated with higher patient BMI (p = 0.024). Normal weight patients had a mean quantitative thymic attenuation of 15.5 Hounsfield unit and overweight patients had a mean quantitative thymic attenuation of -16.4 Hounsfield unit (p = 0.0218). There was a significant association between increasing subcutaneous fat thickness and reduced mean quantitative thymic attenuation (p < 0.0001). There was also a significant difference in subcutaneous fat thickness when comparing qualitatively assessed thymic Grade 0 with grades 2 and 3 (p = 0.027 and 0.001 respectively); and Grade 1 with Grade 3 (p = 0.001). CONCLUSION In patients between 20 and 30 years old, the degree of thymic fatty infiltration is related to BMI. Advances in knowledge: Multidetector CT can assess fatty involution of the thymus gland. This retrospective study demonstrates a relationship between BMI and thymus gland fatty involution. Subjects with increased subcutaneous fat have decreased mean thymus gland attenuation.
Collapse
Affiliation(s)
- Kate A Harrington
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - David S Kennedy
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - Bobby Tang
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - Conor Hickie
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - Emma Phelan
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - William Torreggiani
- 1 Radiology Department, Adelaide and Meath Hospital , Tallaght, Dublin , Ireland
| | - Darragh Halpenny
- 2 Radiology Department, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| |
Collapse
|
142
|
Magnuson AM, Fouts JK, Regan DP, Booth AD, Dow SW, Foster MT. Adipose tissue extrinsic factor: Obesity-induced inflammation and the role of the visceral lymph node. Physiol Behav 2018; 190:71-81. [PMID: 29501838 DOI: 10.1016/j.physbeh.2018.02.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/24/2017] [Accepted: 02/22/2018] [Indexed: 12/17/2022]
Abstract
Obesity-related adverse health consequences occur predominately in individuals with upper body fat distribution commonly associated with increased central adiposity. Visceral adipose tissue accumulation is described to be the greatest driver of obesity-induced inflammation, however evidence also supports that the intestines fundamentally contribute to the development of obesity-induced metabolic disease. The visceral adipose depot shares the same vasculature and lymph drainage as the small intestine. We hypothesize that the visceral lymph node, which drains adipose tissue and the gastrointestinal tract, is central to the exacerbation of systemic pro-inflammation. Male C57BL/6 mice were fed CHOW or high fat diet (HFD) for 7 weeks. At termination the mesenteric depot, visceral lymph node and ileum, jejunum and Peyer's patches were collected. Cytokine concentration was determined in adipose tissue whereas immune cell populations where investigated in the visceral lymph node and intestinal segments by flow cytometry. Visceral adipose tissue and the gastrointestinal tract mutually influence immune cells enclosed within the visceral lymph node. HFD increased visceral lymph node immune cell number. This likely resulted from 1.) an increase in immune cells migration from the small intestines likely from activated dendritic cells that travel to the lymph node and 2.) cytokine effluent from visceral adipose tissue that promoted expansion, survival and retention of pro-inflammatory immune cells. Overall, the visceral lymph node, the immune nexus of visceral adipose tissue and the small intestines, likely plays a fundamental role in exacerbation of systemic pro-inflammation by HFD-induced obesity. The research of Tim Bartness greatly enhanced the understanding of adipose tissue regulation. Studies from his laboratory significantly contributed to our awareness of extrinsic factors that influence body fatness levels. Specifically, the work he produced eloquently demonstrated that adipose tissue was more complex than an insulating storage center; it was connected to our brains via the sympathetic and sensory nervous system. Mapping studies demonstrated that adipose tissue both receives and sends information to the brain. Further, his lab demonstrated that nervous system connections contributed to lipolysis, thermogenesis and adipocyte proliferation and growth. The work of Tim Bartness will continue to influence adipose tissue research. As such, Tim Bartness directly inspired the following research. Adipose tissue extrinsic factors are not limited to the peripheral nervous system. The lymphatic system is an additional extrinsic factor that cross talks with adipose tissue, however its role in this context is under emphasized. Here we begin to elucidate how the lymphatic system may contribute to the comorbidities associated with visceral adipose tissue accumulation.
Collapse
Affiliation(s)
| | | | - Daniel P Regan
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Steve W Dow
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | | |
Collapse
|
143
|
Palmer S, Albergante L, Blackburn CC, Newman TJ. Thymic involution and rising disease incidence with age. Proc Natl Acad Sci U S A 2018; 115:1883-1888. [PMID: 29432166 PMCID: PMC5828591 DOI: 10.1073/pnas.1714478115] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
For many cancer types, incidence rises rapidly with age as an apparent power law, supporting the idea that cancer is caused by a gradual accumulation of genetic mutations. Similarly, the incidence of many infectious diseases strongly increases with age. Here, combining data from immunology and epidemiology, we show that many of these dramatic age-related increases in incidence can be modeled based on immune system decline, rather than mutation accumulation. In humans, the thymus atrophies from infancy, resulting in an exponential decline in T cell production with a half-life of ∼16 years, which we use as the basis for a minimal mathematical model of disease incidence. Our model outperforms the power law model with the same number of fitting parameters in describing cancer incidence data across a wide spectrum of different cancers, and provides excellent fits to infectious disease data. This framework provides mechanistic insight into cancer emergence, suggesting that age-related decline in T cell output is a major risk factor.
Collapse
Affiliation(s)
- Sam Palmer
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom;
- School of Mathematical and Computer Sciences, Heriot-Watt University Malaysia, 62200 Putrajaya, Malaysia
| | - Luca Albergante
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
- Institut Curie, Université de Recherche Paris Sciences et Lettres, Mines ParisTech, INSERM U900, F-75005 Paris, France
| | - Clare C Blackburn
- Medical Research Council Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, EH16 4UU Edinburgh, United Kingdom
| | - T J Newman
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom;
| |
Collapse
|
144
|
Bone marrow lympho-myeloid malfunction in obesity requires precursor cell-autonomous TLR4. Nat Commun 2018; 9:708. [PMID: 29453396 PMCID: PMC5816016 DOI: 10.1038/s41467-018-03145-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/23/2018] [Indexed: 12/21/2022] Open
Abstract
Obesity, a prevalent condition in adults and children, impairs bone marrow (BM) function. However, the underlying mechanisms are unclear. Here, we show that obese mice exhibit poor emergency immune responses in a toll-like receptor 4 (TLR4)-dependent manner. Canonical myeloid genes (Csf1r, Spi1, Runx1) are enhanced, and lymphoid genes (Flt3, Tcf3, Ebf1) are reduced. Using adoptive transfer and mixed BM chimera approaches we demonstrate that myeloid>lymphoid bias arises after 6 weeks of high-fat diet and depends on precursor cell-autonomous TLR4. Further, lean mice exposed to the TLR4 ligand lipopolysaccharide (LPS) at doses similar to that detectable in obese serum recapitulates BM lympho-myeloid alterations. Together, these results establish a mechanistic contribution of BM cell-intrinsic TLR4 to obesity-driven BM malfunction and demonstrate the importance of LPS. Our findings raises important questions about the impact of maternal obesity and endotoxemia to fetal hematopoiesis, as fetal immune precursors are also sensitive to TLR4 signals. Obesity can affect bone marrow cell differentiation and the generation of myeloid and lymphoid cells. Here, the authors show that diet and obesity, as well as low-dose lipopolysaccharide, can alter Toll-like receptor 4 signaling bone marrow cells to skew the myeloid-lymphoid homeostasis in mice.
Collapse
|
145
|
Scully T, Scott CD, Firth SM, Sedger LM, Pintar JE, Twigg SM, Baxter RC. Enhancement of mammary tumour growth by IGFBP-3 involves impaired T cell accumulation. Endocr Relat Cancer 2018; 25:111-122. [PMID: 29217518 DOI: 10.1530/erc-17-0384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/31/2017] [Indexed: 01/08/2023]
Abstract
Epidemiological studies show an association between obesity and poor breast cancer prognosis. We previously demonstrated that global IGFBP-3 deficiency, in IGFBP-3-null mice, resulted in a 50% reduction in mammary tumour growth over 3 weeks relative to tumours in wild-type (WT) C57BL/6 mice. This growth reduction was ameliorated by high fat feeding-induced obesity. This study aimed to examine how IGFBP-3 promotes tumour growth by influencing the immune tumour microenvironment in healthy and obese mice. Syngeneic EO771 cells, which lack detectable IGFBP-3 expression, were grown as orthotopic tumours in WT and IGFBP-3-null C57BL/6 mice placed on either a control chow or a high-fat diet (HFD), and examined by quantitative PCR and immunohistochemistry. In WT mice, increased stromal expression of IGFBP-3 was positively associated with tumour growth, supporting the hypothesis that IGFBP-3 in the microenvironment promotes tumour progression. Examining markers of immune cell subsets, gene expression of Ifng, Cd8a, Cd8b1 and Tnf and CD8 measured by immunohistochemistry were elevated in tumours of IGFBP-3-null mice compared to WT, indicating an accumulation of CD8+ T cells, but this increase was absent if the IGFBP-3-null mice had been exposed to HFD. Expression of these genes was negatively associated with tumour growth. Although similar among groups overall, Nkg2d and Tnfsf10 tumoural expression was associated with decreased tumour growth. Overall, the results of this study provide an immune-based mechanism by which host IGFBP-3 may promote breast tumour growth in the EO771 murine breast cancer model, and suggest that targeting IGFBP-3 might make a novel contribution to immune therapy for breast cancer.
Collapse
Affiliation(s)
- Tiffany Scully
- Hormones and Cancer LaboratoriesKolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Carolyn D Scott
- Hormones and Cancer LaboratoriesKolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Sue M Firth
- Hormones and Cancer LaboratoriesKolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Lisa M Sedger
- School of Life SciencesFaculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - John E Pintar
- Department of Neuroscience and Cell BiologyRutgers Robert Wood Johnson Medical School, New Jersey, USA
| | - Stephen M Twigg
- Charles Perkins CentreSydney Medical School, University of Sydney, New South Wales, Australia
| | - Robert C Baxter
- Hormones and Cancer LaboratoriesKolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
146
|
Aspinall R, Lang PO. Interventions to restore appropriate immune function in the elderly. IMMUNITY & AGEING 2018; 15:5. [PMID: 29416551 PMCID: PMC5785902 DOI: 10.1186/s12979-017-0111-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/28/2017] [Indexed: 01/03/2023]
Abstract
Advanced age is one indicator of likely immune dysfunction. As worldwide, the global population contains progressively more and more older individuals there is likelihood of an increased prevalence and incidence of infectious diseases due to common and emergent pathogens. The resultant increase in mortality and morbidity would be matched by the risk of functional decline and disability. Maintaining immune function at a plateau throughout life may therefore be associated with considerable cost savings. The aim of improving immune function in older individuals may be achieved through considering a therapeutic approach to rejuvenate, stimulate or support the indigenous immune system to perform in a more optimal manner. In terms of cost effectiveness a therapeutic approach may prove difficult because of issues associated with; identifying those who would benefit the most from this treatment, identifying the type of treatment which would suit them and identifying whether the treatment was successful. The alternative of supporting or providing a stronger stimulus through vaccination, whilst more cost effective, may be a more valuable option in the short term. Both approaches will be addressed in this review.
Collapse
Affiliation(s)
- Richard Aspinall
- Rivock Ltd, Bury St Edmunds, UK.,3Anglia Ruskin University, Cambridge, UK
| | - Pierre Olivier Lang
- 2Geriatric and Geriatric Rehabilitation Division, Department of Medicine, University Hospital of Lausanne, Lausanne, Switzerland.,3Anglia Ruskin University, Cambridge, UK
| |
Collapse
|
147
|
Abstract
Solid tumor growth and metastasis require the interaction of tumor cells with the surrounding tissue, leading to a view of tumors as tissue-level phenomena rather than exclusively cell-intrinsic anomalies. Due to the ubiquitous nature of adipose tissue, many types of solid tumors grow in proximate or direct contact with adipocytes and adipose-associated stromal and vascular components, such as fibroblasts and other connective tissue cells, stem and progenitor cells, endothelial cells, innate and adaptive immune cells, and extracellular signaling and matrix components. Excess adiposity in obesity both increases risk of cancer development and negatively influences prognosis in several cancer types, in part due to interaction with adipose tissue cell populations. Herein, we review the cellular and noncellular constituents of the adipose "organ," and discuss the mechanisms by which these varied microenvironmental components contribute to tumor development, with special emphasis on obesity. Due to the prevalence of breast and prostate cancers in the United States, their close anatomical proximity to adipose tissue depots, and their complex epidemiologic associations with obesity, we particularly highlight research addressing the contribution of adipose tissue to the initiation and progression of these cancer types. Obesity dramatically modifies the adipose tissue microenvironment in numerous ways, including induction of fibrosis and angiogenesis, increased stem cell abundance, and expansion of proinflammatory immune cells. As many of these changes also resemble shifts observed within the tumor microenvironment, proximity to adipose tissue may present a hospitable environment to developing tumors, providing a critical link between adiposity and tumorigenesis. © 2018 American Physiological Society. Compr Physiol 8:237-282, 2018.
Collapse
Affiliation(s)
- Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashley M. Fuller
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Liza Makowski
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
148
|
Fujimoto E, Imai A, Utsuyama M, Sato K. Effects of in vitro heat shock on immune cells in diet-induced obese mice. J Therm Biol 2017; 69:124-131. [PMID: 29037372 DOI: 10.1016/j.jtherbio.2017.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022]
Abstract
Obesity has been associated with impaired immune responses and inflammation. The mechanisms underlying these immune disturbances in obesity are not yet clarified. This study investigated the effects of in vitro heat shock (HS) on immune cells from the point of view of thymocyte apoptosis and T-cell mitogen-stimulated splenocyte cytokine production as well as the heat shock protein 70 (HSP70) protein levels in diet-induced obese mice to explore a possible association between the disturbance of T cell immunity and HS response in obesity. Obese mice had increased apoptotic and necrotic thymocytes populations and increased splenocyte cytokine production of both proinflammatory and anti-inflammatory cytokines compared with lean mice. The in vitro HS at 42°C decreased the rate of live cells in thymocytes, and the degree of the decrease was larger in obese mice compared with lean mice. The in vitro HS increased the intracellular and extracellular HSP70 protein levels in thymocytes and splenocytes, while the effects of obesity on the HSP70 protein levels were not obvious. The in vitro HS prior to T cell mitogen stimulation decreased IFN-γ and IL-10 production by mitogen-stimulated splenocytes. This change in cytokine production due to HS was not affected by obesity. The obvious alteration of the HSP70 protein levels and association between cytokine production and the HS response in obesity were not found in this obesity model; however, our results indicate an association between the viability of thymocytes and an altered HS response in obesity and provide evidence that the increase in thymocyte apoptosis and acceleration of thymus involution in obesity could be, in part, due to the alteration of the HS response.
Collapse
Affiliation(s)
- Eka Fujimoto
- Division of Human Development, Graduate School of Human Life Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
| | - Atsuko Imai
- Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
| | - Masanori Utsuyama
- Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
| | - Kazuto Sato
- Division of Human Development, Graduate School of Human Life Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan; Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
149
|
Bharath LP, Ip BC, Nikolajczyk BS. Adaptive Immunity and Metabolic Health: Harmony Becomes Dissonant in Obesity and Aging. Compr Physiol 2017; 7:1307-1337. [PMID: 28915326 DOI: 10.1002/cphy.c160042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue (AT) is the primary energy reservoir organ, and thereby plays a critical role in energy homeostasis and regulation of metabolism. AT expands in response to chronic overnutrition or aging and becomes a major source of inflammation that has marked influence on systemic metabolism. The chronic, sterile inflammation that occurs in the AT during the development of obesity or in aging contributes to onset of devastating diseases such as insulin resistance, diabetes, and cardiovascular pathologies. Numerous studies have shown that inflammation in the visceral AT of humans and animals is a critical trigger for the development of metabolic syndrome. This work underscores the well-supported conclusion that the inflammatory immune response and metabolic pathways in the AT are tightly interwoven by multiple layers of relatively conserved mechanisms. During the development of diet-induced obesity or age-associated adiposity, cells of the innate and the adaptive immune systems infiltrate and proliferate in the AT. Macrophages, which dominate AT-associated immune cells in mouse models of obesity, but are less dominant in obese people, have been studied extensively. However, cells of the adaptive immune system, including T cells and B cells, contribute significantly to AT inflammation, perhaps more in humans than in mice. Lymphocytes regulate recruitment of innate immune cells into AT, and produce cytokines that influence the helpful-to-harmful inflammatory balance that, in turn, regulates organismal metabolism. This review describes inflammation, or more precisely, metabolic inflammation (metaflammation) with an eye toward the AT and the roles lymphocytes play in regulation of systemic metabolism during obesity and aging. © 2017 American Physiological Society. Compr Physiol 7:1307-1337, 2017.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Blanche C Ip
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Molecular Pharmacology, Physiology and Biotechnology, Center of Biomedical Engineering, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
150
|
Frechette DM, Krishnamoorthy D, Pamon T, Chan ME, Patel V, Rubin CT. Mechanical signals protect stem cell lineage selection, preserving the bone and muscle phenotypes in obesity. Ann N Y Acad Sci 2017; 1409:33-50. [PMID: 28891202 DOI: 10.1111/nyas.13442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/19/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
The incidence of obesity is rapidly rising, increasing morbidity and mortality rates worldwide. Associated comorbidities include type 2 diabetes, heart disease, fatty liver disease, and cancer. The impact of excess fat on musculoskeletal health is still unclear, although it is associated with increased fracture risk and a decline in muscular function. The complexity of obesity makes understanding the etiology of bone and muscle abnormalities difficult. Exercise is an effective and commonly prescribed nonpharmacological treatment option, but it can be difficult or unsafe for the frail, elderly, and morbidly obese. Exercise alternatives, such as low-intensity vibration (LIV), have potential for improving musculoskeletal health, particularly in conditions with excess fat. LIV has been shown to influence bone marrow mesenchymal stem cell differentiation toward higher-order tissues (i.e., bone) and away from fat. While the exact mechanisms are not fully understood, recent studies utilizing LIV both at the bench and in the clinic have demonstrated some efficacy. Here, we discuss the current literature investigating the effects of obesity on bone, muscle, and bone marrow and how exercise and LIV can be used as effective treatments for combating the negative effects in the presence of excess fat.
Collapse
Affiliation(s)
- Danielle M Frechette
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - Divya Krishnamoorthy
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - Tee Pamon
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - M Ete Chan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - Vihitaben Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| |
Collapse
|