1
|
Nekrasova AI, Kalashnikova IG, Korobeynikova AV, Ashniev GA, Bobrova MM, Bakoev SY, Petryaikina ES, Nekrasov AS, Zagainova AV, Lukashina MV, Tolkacheva LR, Bobrovnitskii IP, Yudin VS, Keskinov AA, Makarov VV, Yudin SM. Characteristics of the Gut Microbiota Composition of the Arctic Zone Residents in the Far Eastern Region. Biomedicines 2024; 12:2472. [PMID: 39595038 PMCID: PMC11591809 DOI: 10.3390/biomedicines12112472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background. In many studies over the past decade, scientists have made a connection between the composition of gut microbiota and human health. A number of publications have shown that gut bacteria are involved in many metabolic and physiological processes of the organism. The composition of the gut microbiome is unique for each person and is formed under the influence of various factors associated with both the individual characteristics of the body and the characteristics of the environment. Different regional characteristics make it necessary for the body to adapt to certain conditions, including temperature fluctuations. Living in areas with low temperatures, such as the Arctic zone, dictates the need for increased energy consumption, which affects the composition of the gut microbiome. Methods. In our study, an extensive questionnaire was conducted among the participants, where many questions were included about the dietary preferences of the study participants, which allowed them us to further divide them into groups according to their diets. Stool samples were collected from participants from 3 groups: Arctic native, Arctic newcomer and the control group. The next step was the isolation of bacterial DNA and sequencing the 16S rRNA gene. The analysis of the results of the diversity of the intestinal microbiota was carried out both with and without taking into account the dietary preferences of the participants. Results. As a result of comparing the intestinal microbiota obtained from residents of the Arctic zone with the gut microbiota of residents of other regions with a milder climate, significant differences are found. These differences may be related to limited food resources and a reduction in the variety of food products characteristic of this Arctic region. t was also found that representatives of the bacterial families Christensenellaceae and Muribaculaceae dominated the control group, both with traditional nutrition and with a dairy-free diet in comparison with the Arctic groups. The control group was dominated by representatives of the Prevotellaceae, Enterobacteriaceae and Comamonadaceae families compared to the Arctic group (with a traditional diet). The results also show that the number of representatives of the families Desulfovibrionaceae (with traditional diet) and Enterobacteriaceae (with milk-free diet) is growing in the Arctic group. Conclusions. In the course of this work, bacterial families characteristic of people living in the Arc-tic zone of the Far Eastern region of the Russian Federation were identified. Poor diet, difficult climatic conditions, and problems with logistics and medical care can have a strong impact on the health of this population. The main type of diet for the inhabitants of the Arctic is the traditional type of diet. They consume a large number of low-cost products, obtainget animal protein from poultry and canned food, and also eat a small number of fresh vegetables and fruits. Such a diet is due to the social status of the study participants and the climatic and geographical features of the region (difficulties in agriculture). With such a diet, we observe a decrease in representatives of the Christensenellaceae, Muribaculaceae, Eubacteriaceae, and Prevotellaceae families and an increase in representatives of the Enterobacteriaceae and Desulfovibrionaceae families among Arctic residents. This imbalance in the futuremay cause, this population may to develop various diseases in the future, including chronic diseases such as obesity, intestinal dysbiosis, inflammatory bowel diseases, and type 2 diabetes.
Collapse
Affiliation(s)
- Alexandra I. Nekrasova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Irina G. Kalashnikova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Anna V. Korobeynikova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - German A. Ashniev
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Maria M. Bobrova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Sirozhdin Yu. Bakoev
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Ekaterina S. Petryaikina
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Alexander S. Nekrasov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Angelika V. Zagainova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Mariya V. Lukashina
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Larisa R. Tolkacheva
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Igor P. Bobrovnitskii
- Federal State Budgetary Scientific Institution “Institute of General Pathology and Pathophysiology”, Baltiyskaya Str., 8, 125315 Moscow, Russia;
- State Scientific Center, the Russian Federation Institute of Biomedical Problems, the Russian Academy of Sciences, Khoroshevskoe Shosse, 76A, 123007 Moscow, Russia
| | - Vladimir S. Yudin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Anton A. Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Valentin V. Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks”, the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (I.G.K.); (A.V.K.); (G.A.A.); (M.M.B.); (S.Y.B.); (E.S.P.); (A.S.N.); (A.V.Z.); (M.V.L.); (L.R.T.); (V.S.Y.); (A.A.K.); (V.V.M.); (S.M.Y.)
| |
Collapse
|
2
|
Gupta K, George A, Attwood K, Gupta A, Roy AM, Gandhi S, Siromoni B, Singh A, Repasky E, Mukherjee S. Association between Environmental Temperature and Survival in Gastroesophageal Cancers: A Population Based Study. Cancers (Basel) 2023; 16:74. [PMID: 38201502 PMCID: PMC10778299 DOI: 10.3390/cancers16010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Cold stress suppresses antitumor response in animal models, leading to tumor growth. Recent studies have also shown a negative correlation between the average annual temperature (AAT) and cancer incidence. We hypothesized that esophageal cancer (EC) and gastric cancer (GC) patients living in warmer climates have improved survival outcomes than those living in colder climates. METHODS We conducted a retrospective analysis using the Surveillance, Epidemiology, and End Results (SEER) database from 1996 to 2015. We retrieved the National Centers for Environmental Information data to calculate the county-level AAT. Cox multivariate regression models were performed to measure the association between temperature (measured continuously at diagnosis and in 5-degree increments) and OS/DSS, adjusting for variables. All associations were compared at a significance level of 0.05. The OS and DSS were summarized using Kaplan-Meier methods. All statistics were performed using SAS version 9.4 (SAS Institute Inc., Cary, NC, USA). RESULTS A total of 17,408 EC patients were analyzed. The average age of the cohort was 65 years, 79% of which were males and 21% were females. Of them, 61.6% had adenocarcinoma, and 37.6% were squamous. After adjusting for covariates, patients in regions with an AAT > 53.5 °F had an 11% improvement in OS [HR 0.89 (95% CI 0.86-0.92), p < 0.0001] and 13% in DSS [HR 0.87 (95% CI 0.84-0.90), p < 0.0001]. When the temperature was analyzed in 5 °F increments, with each increment, there was a 3% improvement in OS [HR 0.97 (95% CI 0.96-0.98), p < 0.0001] and 4% in DSS [HR 0.96 (95% CI 0.95-0.97), p < 0.0001]. Subgroup analysis of squamous and adenocarcinoma showed similar results. These findings were validated in 20,553 GC patients. After adjusting for covariates, patients in regions with an AAT > 53.5 had a 13% improvement in OS [HR 0.87 (95% CI 0.85-0.90), p < 0.0001] and 14% in DSS [HR 0.86 (95% CI 0.83-0.89), p < 0.0001]. When analyzed in 5 °F increments, with each increment, there was a 4% improvement in OS [HR 0.96 (95% CI 0.952-0.971), p < 0.0001] and 4% in DSS [HR 0.96 (95% CI 0.945-0.965), p < 0.0001]. CONCLUSION We showed for the first time that higher environmental temperatures are associated with significant improvements in OS and DSS in patients with gastro-esophageal cancers, notwithstanding the limitations of a retrospective database analysis. Further confirmatory and mechanistic studies are required to implement specific interventional strategies.
Collapse
Affiliation(s)
- Kush Gupta
- Department of Internal Medicine, Umass Chan Medical School—Baystate, Springfield, MA 01199, USA;
| | - Anthony George
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (K.A.)
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (K.A.)
| | - Ashish Gupta
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| | - Arya Mariam Roy
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| | - Shipra Gandhi
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| | - Beas Siromoni
- School of Health Sciences, University of South Dakota, Vermillion, SD 57069, USA
| | - Anurag Singh
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| | - Elizabeth Repasky
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| | - Sarbajit Mukherjee
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA (A.M.R.)
| |
Collapse
|
3
|
MacDonald CR, Choi JE, Hong CC, Repasky EA. Consideration of the importance of measuring thermal discomfort in biomedical research. Trends Mol Med 2023; 29:589-598. [PMID: 37330365 PMCID: PMC10619709 DOI: 10.1016/j.molmed.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/19/2023]
Abstract
Core temperature stability is the result of a dynamically regulated balance of heat loss and gain, which is not reflected by a simple thermometer reading. One way in which these changes manifest is in perceived thermal comfort, 'feeling too cold' or 'feeling too hot', which can activate stress pathways. Unfortunately, there is surprisingly little preclinical research that tracks changes in perceived thermal comfort in response to either disease progression or various treatments. Without measuring this endpoint, there may be missed opportunities to evaluate disease and therapy outcomes in murine models of human disease. Here, we discuss the possibility that changes in thermal comfort in mice could be a useful and physiologically relevant measure of energy trade-offs required under various physiological or pathological conditions.
Collapse
Affiliation(s)
- Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Jee Eun Choi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
4
|
Ammons DT, MacDonald CR, Chow L, Repasky EA, Dow S. Chronic adrenergic stress and generation of myeloid-derived suppressor cells: Implications for cancer immunotherapy in dogs. Vet Comp Oncol 2023; 21:159-165. [PMID: 36876492 DOI: 10.1111/vco.12891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
Recent studies have highlighted a key role played by the sympathetic nervous system (SNS) and adrenergic stress in mediating immune suppression associated with chronic inflammation in cancer and other diseases. The connection between chronic SNS activation, adrenergic stress and immune suppression is linked in part to the ability of catecholamines to stimulate the bone marrow release and differentiation of myeloid-derived suppressor cells (MDSC). Rodent model studies have revealed an important role for β-adrenergic receptor signalling in suppression of cancer immunity in mice subjected to chronic stresses, including thermal stress. Importantly, therapeutic blockade of beta-adrenergic responses by drugs such as propranolol can partially reverse the generation and differentiation of MDSC, and partly restore tumour immunity. Clinical trials in both humans and dogs with cancer have demonstrated that propranolol blockade can improve responses to radiation therapy, cancer vaccines and immune checkpoint inhibitors. Thus, the SNS stress response has become an important new target to relieve immune suppression in cancer and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Dylan T Ammons
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Microbiology, Immunology, and Pathology, Fort Collins, Colorado, USA
| | - Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Lyndah Chow
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Steven Dow
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
5
|
Wong CP, Branscum AJ, Fichter AR, Sargent J, Iwaniec UT, Turner RT. Cold stress during room temperature housing alters skeletal response to simulated microgravity (hindlimb unloading) in growing female C57BL6 mice. Biochimie 2022:S0300-9084(22)00333-9. [PMID: 36584865 DOI: 10.1016/j.biochi.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 12/29/2022]
Abstract
Laboratory mice are typically housed at temperatures below the thermoneutral zone for the species, resulting in cold stress and premature cancellous bone loss. Furthermore, mice are more dependent upon non-shivering thermogenesis to maintain body temperature during spaceflight, suggesting that microgravity-induced bone loss may be due, in part, to altered thermogenesis. Consequently, we assessed whether housing mice at room temperature modifies the skeletal response to simulated microgravity. This possibility was tested using the hindlimb unloading (HLU) model to mechanically unload femora. Humeri were also assessed as they remain weight bearing during HLU. Six-week-old female C57BL6 (B6) mice were housed at room temperature (22 °C) or near thermoneutral (32 °C) and HLU for 2 weeks. Compared to baseline, HLU resulted in cortical bone loss in femur, but the magnitude of reduction was greater in mice housed at 22 °C. Cancellous osteopenia in distal femur (metaphysis and epiphysis) was noted in HLU mice housed at both temperatures. However, bone loss occurred at 22 °C, whereas the bone deficit at 32 °C was due to failure to accrue bone. HLU resulted in cortical and cancellous bone deficits (compared to baseline) in humeri of mice housed at 22 °C. In contrast, fewer osteopenic changes were detected in mice housed at 32 °C. These findings support the hypothesis that environmental temperature alters the skeletal response to HLU in growing female mice in a bone compartment-specific manner. Taken together, species differences in thermoregulation should be taken into consideration when interpreting the skeletal response to simulated microgravity.
Collapse
Affiliation(s)
- Carmen P Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Adam J Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Aidan R Fichter
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Jennifer Sargent
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331 USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA; Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA; Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
6
|
Liu Y, Shi H, Hu Y, Yao R, Liu P, Yang Y, Li S. RNA binding motif protein 3 (RBM3) promotes protein kinase B (AKT) activation to enhance glucose metabolism and reduce apoptosis in skeletal muscle of mice under acute cold exposure. Cell Stress Chaperones 2022; 27:603-618. [PMID: 36149580 PMCID: PMC9672220 DOI: 10.1007/s12192-022-01297-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/01/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023] Open
Abstract
The main danger of cold stress to animals in cold regions is systemic metabolic changes and protein synthesis inhibition. RBM3, an exceptional cold shock protein, is rapidly upregulated in response to hypothermia to resist the adverse effects of cold stress. However, the mechanism of the protective effect and the rapid upregulation of RBM3 remains unclear. O-GlcNAcylation, an atypical O-glycosylation, is precisely regulated only by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) and participates in the signal transduction of multiple cellular stress responses as a "stress and nutrition receptor." Therefore, our study aimed to explore the mechanism of RBM3 regulating glucose metabolism and promoting survival in skeletal muscle under acute cold exposure. Meanwhile, our study verifies whether O-GlcNAcylation mediated by OGT rapidly upregulates RBM3. The blood and skeletal muscle of mice were collected at the end of cold exposure treatment for 0, 2, and 4 h. Changes in levels of RBM3, AKT, glycolysis apoptosis, and OGT were measured. The results show that acute cold exposure upregulated RBM3, OGT, and AKT phosphorylation and increased energy consumption, which enhanced glycolysis and prevent apoptosis. In the 32 °C mild hypothermia model in vitro, overexpression of RBM3 enhanced AKT phosphorylation. Meanwhile, inactivation of AKT by wortmannin resulted in increased apoptosis and decreased glucose metabolism in skeletal muscle under acute cold exposure. In addition, OGT-mediated O-GlcNAcylation of p65 was confirmed in mouse myoblast cell line (C2C12) cells at mild hypothermia. O-GlcNAcylation level affected p65 activity and nuclear translocation. Compared with wild type (WT) mice, RBM3 and p65 phosphorylation were decreased in specific skeletal muscle Ogt (KO) mice, whereas AKT phosphorylation, glycolysis, and apoptosis were increased. Taken together, O-GlcNAcylation of p65 upregulates RBM3 to promote AKT phosphorylation, enhance glucose metabolism, and reduce apoptosis in skeletal muscle of mice under acute cold exposure.
Collapse
Affiliation(s)
- Yang Liu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Hongzhao Shi
- Department of Animal Engineering, Yangling Vocational & Technical College, Xianyang, 712199, People's Republic of China
| | - Yajie Hu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Ruizhi Yao
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, 028000, People's Republic of China
| | - Peng Liu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Yuying Yang
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Shize Li
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China.
| |
Collapse
|
7
|
Nunamaker EA, Reynolds PS. 'Invisible actors'-How poor methodology reporting compromises mouse models of oncology: A cross-sectional survey. PLoS One 2022; 17:e0274738. [PMID: 36264974 PMCID: PMC9584398 DOI: 10.1371/journal.pone.0274738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/28/2022] [Indexed: 11/05/2022] Open
Abstract
The laboratory mouse is a key player in preclinical oncology research. However, emphasis of techniques reporting at the expense of critical animal-related detail compromises research integrity, animal welfare, and, ultimately, the translation potential of mouse-based oncology models. To evaluate current reporting practices, we performed a cross-sectional survey of 400 preclinical oncology studies using mouse solid-tumour models. Articles published in 2020 were selected from 20 journals that specifically endorsed the ARRIVE (Animal Research: Reporting of In Vivo Experiments) preclinical reporting guidelines. We assessed reporting compliance for 22 items in five domains: ethical oversight assurance, animal signalment, husbandry, welfare, and euthanasia. Data were analysed using hierarchical generalised random-intercept models, clustered on journal. Overall, reporting of animal-related items was poor. Median compliance over all categories was 23%. There was little or no association between extent of reporting compliance and journal or journal impact factor. Age, sex, and source were reported most frequently, but verifiable strain information was reported for <10% of studies. Animal husbandry, housing environment, and welfare items were reported by <5% of studies. Fewer than one in four studies reported analgesia use, humane endpoints, or an identifiable method of euthanasia. Of concern was the poor documentation of ethical oversight information. Fewer than one in four provided verifiable approval information, and almost one in ten reported no information, or information that was demonstrably false. Mice are the "invisible actors" in preclinical oncology research. In spite of widespread endorsement of reporting guidelines, adherence to reporting guidelines on the part of authors is poor and journals fail to enforce guideline reporting standards. In particular, the inadequate reporting of key animal-related items severely restricts the utility and translation potential of mouse models, and results in research waste. Both investigators and journals have the ethical responsibility to ensure animals are not wasted in uninformative research.
Collapse
Affiliation(s)
- Elizabeth A. Nunamaker
- Animal Care Services, University of Florida, Gainesville, Florida, United States of America
| | - Penny S. Reynolds
- Department of Anesthesiology, Statistics in Anesthesiology Research (STAR) Core, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
8
|
Liu M, Zou X, Fu M, Bai X, Zhao Y, Chen X, Wang X, Wang P, Huang S. Mild cold stress specifically disturbs clustering movement of DFCs and sequential organ left-right patterning in zebrafish. Front Cell Dev Biol 2022; 10:952844. [PMID: 36211472 PMCID: PMC9539758 DOI: 10.3389/fcell.2022.952844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
In poikilothermic animals, the distinct acclimatization ability of different organs has been previously addressed, while the tissue-specific role of cold stress in early development is largely unknown. In this study, we discovered that despite its role in delaying embryonic development, mild cold stress (22°C) does not disturb multiple-organ progenitor specification, but does give rise to organ left-right (LR) patterning defects. Regarding the mechanism, the data showed that mild cold stress downregulated the expression of cell-adhesion genes cdh1 and cdh2 during gastrulation, especially in dorsal forerunner cells (DFCs), which partially disturbed the clustering movement of DFCs, Kupffer’s vesicle (KV) morphogenesis, and ciliogenesis. As a result, the defects of KV/cilia disrupted asymmetric nodal signaling and subsequent heart and liver LR patterning. In conclusion, our data novelly identified that, in early development, DFCs are more sensitive to mild cold stress, and mild cold stress repressed the expression of cell adhesion-related gene cdh1 and cdh2. This role partially disturbed the clustering movement of DFCs, which resulted in defective KV/cilia development and sequential organ LR patterning defects.
Collapse
Affiliation(s)
- Min Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- Department of Cardiology, the First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Xinyu Zou
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Mao Fu
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xinping Bai
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Yongyan Zhao
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xin Chen
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Xiaoyu Wang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Peijian Wang
- Department of Cardiology, the First Affiliated Hospital, Chengdu Medical College, Chengdu, China
- *Correspondence: Peijian Wang, ; Sizhou Huang,
| | - Sizhou Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- *Correspondence: Peijian Wang, ; Sizhou Huang,
| |
Collapse
|
9
|
Liu T, Guo Y, Lu C, Cai C, Gao P, Cao G, Li B, Guo X, Yang Y. Effect of Different Pig Fecal Microbiota Transplantation on Mice Intestinal Function and Microbiota Changes During Cold Exposure. Front Vet Sci 2022; 9:805815. [PMID: 35498721 PMCID: PMC9044030 DOI: 10.3389/fvets.2022.805815] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/09/2022] [Indexed: 12/27/2022] Open
Abstract
Cold stress influences intestinal processes, causing physiological and immunological responses in animals. Intestinal microbiota participates in maintaining the stability of the intestinal environment. However, phenotypic characteristics and the effects of porcine microbiota changes under cold conditions remain poorly understood. Here, the fecal microbiota of cold tolerant breed (Mashen) and cold sensitive breed (Duroc-Landrace-Yorkshire) was transferred to germ-free mice, respectively. After a cold exposure (4°C) for 21 days, intestinal function and microbe changes of mice were explored. The results showed that Mashen pigs microbiota transplantation made the body temperature of the mice stable, in which the fat weight and expression of uncoupling protein 1 (UCP1), carnitine palmitoyltransferase 1B (Cpt1b), and Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) were significantly higher (P < 0.05) than those of the control group. The results of intestinal structure and expression of serum inflammatory factors showed that fecal microbiota transplantation (FMT) mice have more intact intestinal structure and high expression of proinflammatory factor such as interleukin-4 (IL-4). The study of mice fecal microbiome characterized via 16S rRNA sequencing found that pig microbiota transplantation changed the abundance of Firmicutes. In addition, it identified discriminative features of Firmicutes in the microbiota between two breeds of pig, in which Clostridiaceae were enriched in the microbiota community of Mashen pig and Coriobacteriales were significantly (P < 0.05) enriched in the Duroc-Landrace-Yorkshire pig microbiota transplantation group based on linear discriminant analysis effect size (LEfSe) analysis. Finally, we found that the content of propionic acid and butyric acid in rectal contents significantly changed and the abundances of Clostridium and Lachnospira showed significant correlations with changes in short-chain fatty acids. The results suggest that pig fecal microbiota transplantation can alleviate the changes in physiological and biochemical indicators in mice caused by cold exposure. Mice have gut microbes altered and improved gut barrier function via fecal microbiota transplantation in pigs.
Collapse
|
10
|
Gregg C. Starvation and Climate Change—How to Constrain Cancer Cell Epigenetic Diversity and Adaptability to Enhance Treatment Efficacy. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.693781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Advanced metastatic cancer is currently not curable and the major barrier to eliminating the disease in patients is the resistance of subpopulations of tumor cells to drug treatments. These resistant subpopulations can arise stochastically among the billions of tumor cells in a patient or emerge over time during therapy due to adaptive mechanisms and the selective pressures of drug therapies. Epigenetic mechanisms play important roles in tumor cell diversity and adaptability, and are regulated by metabolic pathways. Here, I discuss knowledge from ecology, evolution, infectious disease, species extinction, metabolism and epigenetics to synthesize a roadmap to a clinically feasible approach to help homogenize tumor cells and, in combination with drug treatments, drive their extinction. Specifically, cycles of starvation and hyperthermia could help synchronize tumor cells and constrain epigenetic diversity and adaptability by limiting substrates and impairing the activity of chromatin modifying enzymes. Hyperthermia could also help prevent cancer cells from entering dangerous hibernation-like states. I propose steps to a treatment paradigm to help drive cancer extinction that builds on the successes of fasting, hyperthermia and immunotherapy and is achievable in patients. Finally, I highlight the many unknowns, opportunities for discovery and that stochastic gene and allele level epigenetic mechanisms pose a major barrier to cancer extinction that warrants deeper investigation.
Collapse
|
11
|
Carlson PM, Mohan M, Rodriguez M, Subbotin V, Sun CX, Patel RB, Birstler J, Hank JA, Rakhmilevich AL, Morris ZS, Erbe AK, Sondel PM. Depth of tumor implantation affects response to in situ vaccination in a syngeneic murine melanoma model. J Immunother Cancer 2021; 9:e002107. [PMID: 33858849 PMCID: PMC8055108 DOI: 10.1136/jitc-2020-002107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 01/15/2023] Open
Abstract
An important component of research using animal models is ensuring rigor and reproducibility. This study was prompted after two experimenters performing virtually identical studies obtained different results when syngeneic B78 murine melanoma cells were implanted into the skin overlying the flank and treated with an in situ vaccine (ISV) immunotherapy. Although both experimenters thought they were using identical technique, we determined that one was implanting the tumors intradermally (ID) and the other was implanting them subcutaneously (SC). Though the baseline in vivo immunogenicity of tumors can depend on depth of their implantation, the response to immunotherapy as a function of tumor depth, particularly in immunologically 'cold' tumors, has not been well studied. The goal of this study was to evaluate the difference in growth kinetics and response to immunotherapy between identically sized melanoma tumors following ID versus SC implantation. We injected C57BL/6 mice with syngeneic B78 melanoma cells either ID or SC in the flank. When tumors reached 190-230 mm3, they were grouped into a 'wave' and treated with our previously published ISV regimen (12 Gy local external beam radiation and intratumoral hu14.18-IL2 immunocytokine). Physical examination demonstrated that ID-implanted tumors were mobile on palpation, while SC-implanted tumors became fixed to the underlying fascia. Histologic examination identified a critical fascial layer, the panniculus carnosus, which separated ID and SC tumors. SC tumors reached the target tumor volume significantly faster compared with ID tumors. Most ID tumors exhibited either partial or complete response to this immunotherapy, whereas most SC tumors did not. Further, the 'mobile' or 'fixed' phenotype of tumors predicted response to therapy, regardless of intended implantation depth. These findings were then extended to additional immunotherapy regimens in four separate tumor models. These data indicate that the physical 'fixed' versus 'mobile' characterization of the tumors may be one simple method of ensuring homogeneity among implanted tumors prior to initiation of treatment. Overall, this short report demonstrates that small differences in depth of tumor implantation can translate to differences in response to immunotherapy, and proposes a simple physical examination technique to ensure consistent tumor depth when conducting implantable tumor immunotherapy experiments.
Collapse
Affiliation(s)
- Peter M Carlson
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Manasi Mohan
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew Rodriguez
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vladimir Subbotin
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Claire X Sun
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ravi B Patel
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jen Birstler
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amy K Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Ishizuka S. Do dominant monkeys gain more warmth? Number of physical contacts and spatial positions in huddles for male Japanese macaques in relation to dominance rank. Behav Processes 2021; 185:104317. [PMID: 33417930 DOI: 10.1016/j.beproc.2021.104317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 12/22/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023]
Abstract
Animals show various forms of behavioral thermoregulation to minimize cold stress. Given that higher dominance rank is often associated with increased fitness in group-living animals, higher-ranking individuals may also benefit from better access to thermally optimal spatial positions within huddles. This study examined the association between dominance rank and the potential thermoregulatory benefits of huddling behavior in Japanese macaques (Macaca fuscata) inhabiting Shodoshima Island, which form exceptionally large huddles. I photographed monkey huddles, and analyzed the number of individuals that males were in contact with and males' spatial positons in huddles. Higher-ranking males were significantly more likely to be in contact with larger numbers of individuals in huddles. Higher-ranking males occupied non-peripheral positions in huddles more often than lower-ranking males, which put them in contact with larger numbers of individuals. These results suggest that high dominance rank may confer potential thermal advantages on male Japanese macaques. The mechanism for this is likely that the highest-ranking male often intrude in already-formed huddles, although such behaviors of males were not quantitatively assessed. This study contributes to a better understanding of the mechanisms of cold adaptation in relation to dominance rank in group-living animals.
Collapse
Affiliation(s)
- Shintaro Ishizuka
- Primate Research Institute, Kyoto University, Japan; Japan Society for Promotion of Science, Japan.
| |
Collapse
|
13
|
Vialard F, Olivier M. Thermoneutrality and Immunity: How Does Cold Stress Affect Disease? Front Immunol 2020; 11:588387. [PMID: 33329571 PMCID: PMC7714907 DOI: 10.3389/fimmu.2020.588387] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
One of the major challenges the scientific community faces today is the lack of translational data generated from mouse trials for human health application. Housing temperature-dependent chronic cold stress in laboratory rodents is one of the key factors contributing to lack of translatability because it reveals major metabolic differences between humans and rodents. While humans tend to operate at temperatures within their thermoneutral zone, most laboratory rodents are housed at temperatures below this zone and have an increased energy demand to generate heat. This has an impact on the immune system of mice and thus affects results obtained using murine models of human diseases. A limited number of studies and reviews have shown that results obtained on mice housed at thermoneutrality were different from those obtained from mice housed in traditional housing conditions. Most of those studies, focused on obesity and cancer, found that housing mice at thermoneutrality changed the outcomes of the diseases negatively and positively, respectively. In this review, we describe how thermoneutrality impacts the immune system of rodents generally and in the context of different disease models. We show that thermoneutrality exacerbates cardiovascular and auto-immune diseases; alleviates asthma and Alzheimer’s disease; and, changes gut microbiome populations. We also show that thermoneutrality can have exacerbating or alleviating effects on the outcome of infectious diseases. Thus, we join the call of others in this field to urge researchers to refine murine models of disease and increase their translational capacity by considering housing at thermoneutrality for trials involving rodents.
Collapse
Affiliation(s)
- Fiorella Vialard
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
14
|
Lee VK, David JM, Huerkamp MJ. Micro- and Macroenvironmental Conditions and Stability of Terrestrial Models. ILAR J 2020; 60:120-140. [PMID: 33094820 DOI: 10.1093/ilar/ilaa013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 01/15/2023] Open
Abstract
Environmental variables can have profound effects on the biological responses of research animals and the outcomes of experiments dependent on them. Some of these influences are both predictable and unpredictable in effect, many are challenging to standardize, and all are influenced by the planning and conduct of experiments and the design and operation of the vivarium. Others are not yet known. Within the immediate environment where the research animal resides, in the vivarium and in transit, the most notable of these factors are ambient temperature, relative humidity, gaseous pollutant by-products of animal metabolism and physiology, dust and particulates, barometric pressure, electromagnetic fields, and illumination. Ambient temperatures in the animal housing environment, in particular those experienced by rodents below the thermoneutral zone, may introduce degrees of stress and thermoregulatory compensative responses that may complicate or invalidate study measurements across a broad array of disciplines. Other factors may have more subtle and specific effects. It is incumbent on scientists designing and executing experiments and staff responsible for animal husbandry to be aware of, understand, measure, systematically record, control, and account for the impact of these factors on sensitive animal model systems to ensure the quality and reproducibility of scientific studies.
Collapse
Affiliation(s)
- Vanessa K Lee
- Department of Pathology and Laboratory Medicine and Division of Animal Resources, School of Medicine, Emory University, Atlanta, Georgia
| | - John M David
- Translational Medicine Department, Vertex Pharmaceuticals, Boston, Massachusetts
| | - Michael J Huerkamp
- Department of Pathology and Laboratory Medicine and Division of Animal Resources, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
15
|
O'Brien CM, Meng H, Shmuylovich L, Carpenter J, Gogineni P, Zhang H, Bishop K, Mondal SB, Sudlow GP, Bethea C, Bethea C, Achilefu S. Focal dynamic thermal imaging for label-free high-resolution characterization of materials and tissue heterogeneity. Sci Rep 2020; 10:12549. [PMID: 32724184 PMCID: PMC7387563 DOI: 10.1038/s41598-020-69362-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/10/2020] [Indexed: 11/30/2022] Open
Abstract
Evolution from static to dynamic label-free thermal imaging has improved bulk tissue characterization, but fails to capture subtle thermal properties in heterogeneous systems. Here, we report a label-free, high speed, and high-resolution platform technology, focal dynamic thermal imaging (FDTI), for delineating material patterns and tissue heterogeneity. Stimulation of focal regions of thermally responsive systems with a narrow beam, low power, and low cost 405 nm laser perturbs the thermal equilibrium. Capturing the dynamic response of 3D printed phantoms, ex vivo biological tissue, and in vivo mouse and rat models of cancer with a thermal camera reveals material heterogeneity and delineates diseased from healthy tissue. The intuitive and non-contact FDTI method allows for rapid interrogation of suspicious lesions and longitudinal changes in tissue heterogeneity with high-resolution and large field of view. Portable FDTI holds promise as a clinical tool for capturing subtle differences in heterogeneity between malignant, benign, and inflamed tissue.
Collapse
Affiliation(s)
- Christine M O'Brien
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Hongyu Meng
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Leonid Shmuylovich
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Julia Carpenter
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Praneeth Gogineni
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Haini Zhang
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Kevin Bishop
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Suman B Mondal
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Gail P Sudlow
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA
| | - Cheryl Bethea
- Quantum Technology Consultants, Inc., 8 Grist Mill Lane, Franklin Park, NJ, 08823, USA
| | - Clyde Bethea
- Quantum Technology Consultants, Inc., 8 Grist Mill Lane, Franklin Park, NJ, 08823, USA
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., Couch Biomedical Research Building, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
| |
Collapse
|
16
|
Bandyopadhayaya S, Ford B, Mandal CC. Cold-hearted: A case for cold stress in cancer risk. J Therm Biol 2020; 91:102608. [PMID: 32716858 DOI: 10.1016/j.jtherbio.2020.102608] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/25/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
A negative correlation exists between environmental temperature and cancer risk based on both epidemiological and statistical analyses. Previously, cold stress was reported to be an effective cause of tumorigenesis. Several studies have demonstrated that cold temperature serves as a potential risk factor in cancer development. Most recently, a link was demonstrated between the effects of extreme cold climate on cancer incidence, pinpointing its impact on tumour suppressor genes by causing mutation. The underlying mechanism behind cold stress and its association with tumorigenesis is not well understood. Hence, this review intends to shed light on the role of associated factors, genetic and/or non-genetic, which are modulated by cold temperature, and eventually influence tumorigenic potential. While scrutinizing the effect of cold exposure on the body, the expression of certain genes, e.g. uncoupled proteins and heat-shock proteins, were elevated. Biological chemicals such as norepinephrine, thyroxine, and cholesterol were also elevated. Brown adipose tissue, which plays an essential role in thermogenesis, displayed enhanced activity upon cold exposure. Adaptive measures are utilized by the body to tolerate the cold, and in doing so, invites both epigenetic and genetic changes. Unknowingly, these adaptive strategies give rise to a lethal outcome i.e., genesis of cancer. Concisely, this review attempts to draw a link between cold stress, genetic and epigenetic changes, and tumorigenesis and aspires to ascertain the mechanism behind cold temperature-mediated cancer risk.
Collapse
Affiliation(s)
| | - Bridget Ford
- Department of Biology, University of the Incarnate Word, San Antonio, TX, 78209, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, 305817, India.
| |
Collapse
|
17
|
Abstract
Regulatory guidelines mandate housing for laboratory mice at temperatures below their thermoneutral zone, creating chronic cold stress. However, increases in housing temperature could alter immune responses. We hypothesized housing mice at temperatures within their thermoneutral zone would improve sepsis survival and alter immune responses. Male C57BL/6 mice were housed at 22°C or 30°C after cecal ligation and puncture (CLP) for 10 days. Survival of mice housed at 30°C (78%) after CLP was significantly increased compared with mice housed at 22°C (40%). Experimental groups were repeated with mice euthanized at 0, 12, 24, and 48 h post-surgery to examine select immune parameters. Raising housing temperature minimally altered systemic, peritoneal, or splenic cell counts. However, IL-6 levels in plasma and peritoneal lavage fluid were significantly lower at 12 h post-surgery in mice housed at 30°C compared with 22°C. Bacterial colony counts from peritoneal lavage fluid were significantly lower in mice housed at 30°C and in vivo studies suggested this was the result of increased phagocytosis by neutrophils. As previously demonstrated, adoptive transfer of fibrocytes significantly increased sepsis survival compared with saline at 22°C. However, there was no additive effect when adoptive transfer was performed at 30°C. Overall, the results demonstrated that thermoneutral housing improves survival after CLP by increasing local phagocytic activity and technical revisions may be necessary to standardize the severity of the model across different housing temperatures. These findings stress the pronounced impact housing temperature has on the CLP model and the importance of reporting housing temperature.
Collapse
|
18
|
Barbee RW, Turner PV. Incorporating Laboratory Animal Science into Responsible Biomedical Research. ILAR J 2019; 60:9-16. [DOI: 10.1093/ilar/ilz017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
Abstract
Biomedical research has made great strides in the past century leading to rapid advances in human life expectancy, all derived from improved understanding, prevention, and treatment of many diseases and conditions. Research involving laboratory animals has played a significant role in this medical progress. However, there continues to be controversy surrounding the use of animals in research, and animal models have been questioned regarding their relevance to human conditions. While research fraud and questionable research practices could potentially contribute to this problem, we argue that a relative ignorance of laboratory animal science has contributed to the “uncontrolled vivarium experiment” that runs parallel to the more controlled scientific experiment. Several variables are discussed, including husbandry, animal environment, social housing, and more, that can contribute to this uncontrolled experiment, and that can simultaneously decrease quality of life for rodent test subjects when ignored. An argument is put forward that laboratory animal veterinarians and scientists can and should play an important role in better controlling such variables. Similarly, the laboratory animal veterinarian and scientist should play an important role in responsible science by addressing complex interdisciplinary challenges.
Collapse
Affiliation(s)
- R Wayne Barbee
- Virginia Commonwealth University, Office of Research and Innovation
| | - Patricia V Turner
- Charles River Laboratories Inc., Global Animal Welfare & Training, University of Guelph Pathobiology
| |
Collapse
|
19
|
Radaelli E, Santagostino SF, Sellers RS, Brayton CF. Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research. ILAR J 2019; 59:211-246. [PMID: 31197363 PMCID: PMC7114723 DOI: 10.1093/ilar/ily026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.
Collapse
Affiliation(s)
- Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara F Santagostino
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California
| | | | - Cory F Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
Hylander BL, Gordon CJ, Repasky EA. Manipulation of Ambient Housing Temperature To Study the Impact of Chronic Stress on Immunity and Cancer in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:631-636. [PMID: 30670578 DOI: 10.4049/jimmunol.1800621] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
Mice are the preeminent research organism in which to model human diseases and study the involvement of the immune response. Rapidly accumulating evidence indicates a significant involvement of stress hormones in cancer progression, resistance to therapies, and suppression of immune responses. As a result, there has been a concerted effort to model human stress in mice. In this article, we discuss recent literature showing how mice in research facilities are chronically stressed at baseline because of environmental factors. Focusing on housing temperature, we suggest that the stress of cool housing temperatures contributes to the impact of other imposed experimental stressors and therefore has a confounding effect on mouse stress models. Furthermore, we propose that manipulation of housing temperature is a useful approach for studying the impact of chronic stress on disease and the immune response and for testing therapeutic methods of reducing the negative effects of chronic stress.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Christopher J Gordon
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| |
Collapse
|
21
|
Gassen J, Proffitt Leyva RP, Mengelkoch S, White JD, Peterman JL, Prokosch ML, Bradshaw HK, Eimerbrink MJ, Corrigan EK, Cheek DJ, Boehm GW, Hill SE. Day length predicts investment in human immune function: Shorter days yield greater investment. Psychoneuroendocrinology 2019; 107:141-147. [PMID: 31128570 DOI: 10.1016/j.psyneuen.2019.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 11/25/2022]
Abstract
Winter is characterized by stressful conditions which compromise health and render animals more vulnerable to infection and illness than during other times of the year. Organisms are hypothesized to adapt to these seasonal stressors by increasing investment in immune function in response to diminished photoperiod duration. Here, we examined this hypothesis in a sample of healthy human participants. Using several functional immune assays in vitro, as well as by utilizing measures of in vivo proinflammatory cytokine levels, we predicted that shorter day length would be associated with greater investment in immunological function. Results revealed that shorter days predicted significant upregulation of several facets of immune function, including natural killer cell cytotoxicity, peripheral blood mononuclear cell proliferation (in response to, and in the absence of stimulation), and plasma levels of interleukin-6, as well as lower rates of Staphylococcus aureus growth in serum ex vivo. Further, consistent with the hypothesis that these trade-offs would be offset by decreased investment in mating effort, shorter day length also predicted lower levels of total testosterone in men. These results suggest that ambient photoperiod may be a powerful regulator of human immunological activity, providing some of the first evidence of seasonal changes in multiple facets of human immune function.
Collapse
Affiliation(s)
- Jeffrey Gassen
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States.
| | - Randi P Proffitt Leyva
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Summer Mengelkoch
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Jordon D White
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Julia L Peterman
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Marjorie L Prokosch
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Hannah K Bradshaw
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Micah J Eimerbrink
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Emily K Corrigan
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Dennis J Cheek
- Texas Christian University, Harris College of Nursing and Health Sciences, 2800 W Bowie St, Fort Worth, TX 76109, United States
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| | - Sarah E Hill
- Texas Christian University, Department of Psychology, 2955 S University Dr, Fort Worth, TX 76109, United States
| |
Collapse
|
22
|
Hankenson FC, Marx JO, Gordon CJ, David JM. Effects of Rodent Thermoregulation on Animal Models in the Research Environment. Comp Med 2018; 68:425-438. [PMID: 30458902 DOI: 10.30802/aalas-cm-18-000049] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To best promote animal wellbeing and the efficacy of biomedical models, scientific, husbandry, and veterinary professionals must consider the mechanisms, influences, and outcomes of rodent thermoregulation in contemporary research environments. Over the last 2 decades, numerous studies have shown that laboratory mice and rats prefer temperatures that are several degrees warmer than the environments in which they typically are housed within biomedical facilities. Physiologic changes to rodents that are cage-housed under standard temperatures (20 to 26 °C) are attributed to 'cold stress' and include alterations in metabolism, cardiovascular parameters, respiration, and immunologic function. This review article describes common behavioral and physiologic adaptations of laboratory mice and rats to cold stress within modern vivaria, with emphasis on environmental enrichment and effects of anesthesia and procedural support efforts. In addition, potential interventions and outcomes for rodents are presented, relative to the importance of repeating and reproducing experiments involving laboratory rodent research models of human disease.
Collapse
Affiliation(s)
- F Claire Hankenson
- Campus Animal Resources, Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - James O Marx
- University Laboratory Animal Resources, Department of Pathobiology, School of Veterinary Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher J Gordon
- Toxicity Assessment Division, Neurotoxicology Branch, United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - John M David
- Comparative Medicine, Pfizer, La Jolla, California, USA
| |
Collapse
|
23
|
Abstract
The mitochondrial hypothesis of aging evolved from the rate-of-living theory. That theory posited that the rate of aging was largely determined by the rate of energy expenditure. The mechanistic link between energy expenditure and aging was hypothesized to be oxidative stress. As both energy expenditure and reactive oxygen species (ROS) centered on the mitochondria that organelle became a central focus of aging research. Until about the turn of the 21st century available evidence largely supported the efficiency of mitochondrial function as a key contributor to aging. However as methods for investigating mitochondrial oxidant production and tissue level oxidative damage improved, evidentiary support for the theory weakened. Recently, direct disruption of mitochondrial function has been shown not to shorten life or health as expected, but in many cases in multiple laboratory species disrupted mitochondrial function has lengthened life, sometimes without apparent tradeoffs. Does this mean that mitochondrial function plays no role in aging as had been posited for many years? One key consideration is that experiments under laboratory conditions can be misleading about physiological processes that occur in the uncertain conditions of nature. Before we discard the mitochondrial hypothesis of aging, more field experiments targeted at that hypothesis need to be performed. Fortunately, emerging technology is making such experiment more possible than ever before.
Collapse
Affiliation(s)
- Steven N Austad
- Department of Biology, University of Alabama at Birmingham, 1720 Second Avenue South, CH 464, Birmingham, AL 35294-1170, USA
| |
Collapse
|
24
|
Qiao G, Chen M, Bucsek MJ, Repasky EA, Hylander BL. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front Immunol 2018; 9:164. [PMID: 29479349 PMCID: PMC5812031 DOI: 10.3389/fimmu.2018.00164] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
An immune response must be tightly controlled so that it will be commensurate with the level of response needed to protect the organism without damaging normal tissue. The roles of cytokines and chemokines in orchestrating these processes are well known, but although stress has long been thought to also affect immune responses, the underlying mechanisms were not as well understood. Recently, the role of nerves and, specifically, the sympathetic nervous system, in regulating immune responses is being revealed. Generally, an acute stress response is beneficial but chronic stress is detrimental because it suppresses the activities of effector immune cells while increasing the activities of immunosuppressive cells. In this review, we first discuss the underlying biology of adrenergic signaling in cells of both the innate and adaptive immune system. We then focus on the effects of chronic adrenergic stress in promoting tumor growth, giving examples of effects on tumor cells and immune cells, explaining the methods commonly used to induce stress in preclinical mouse models. We highlight how this relates to our observations that mandated housing conditions impose baseline chronic stress on mouse models, which is sufficient to cause chronic immunosuppression. This problem is not commonly recognized, but it has been shown to impact conclusions of several studies of mouse physiology and mouse models of disease. Moreover, the fact that preclinical mouse models are chronically immunosuppressed has critical ramifications for analysis of any experiments with an immune component. Our group has found that reducing adrenergic stress by housing mice at thermoneutrality or treating mice housed at cooler temperatures with β-blockers reverses immunosuppression and significantly improves responses to checkpoint inhibitor immunotherapy. These observations are clinically relevant because there are numerous retrospective epidemiological studies concluding that cancer patients who were taking β-blockers have better outcomes. Clinical trials testing whether β-blockers can be repurposed to improve the efficacy of traditional and immunotherapies in patients are on the horizon.
Collapse
Affiliation(s)
- Guanxi Qiao
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Minhui Chen
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark J. Bucsek
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Elizabeth A. Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bonnie L. Hylander
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
25
|
Appenheimer MM, Evans SS. Temperature and adaptive immunity. HANDBOOK OF CLINICAL NEUROLOGY 2018; 156:397-415. [DOI: 10.1016/b978-0-444-63912-7.00024-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
26
|
Ehlers A, Xie W, Agapov E, Brown S, Steinberg D, Tidwell R, Sajol G, Schutz R, Weaver R, Yu H, Castro M, Bacharier LB, Wang X, Holtzman MJ, Haspel JA. BMAL1 links the circadian clock to viral airway pathology and asthma phenotypes. Mucosal Immunol 2018; 11:97-111. [PMID: 28401936 PMCID: PMC5638664 DOI: 10.1038/mi.2017.24] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 03/01/2017] [Indexed: 02/04/2023]
Abstract
Patients with asthma experience circadian variations in their symptoms. However it remains unclear how specific aspects of this common airway disease relate to clock genes, which are critical to the generation of circadian rhythms in mammals. Here, we used a viral model of acute and chronic airway disease to examine how circadian clock disruption affects asthmatic lung phenotypes. Deletion of the core clock gene bmal1 or environmental disruption of circadian function by jet lag exacerbated acute viral bronchiolitis caused by Sendai virus (SeV) and influenza A virus in mice. Post-natal deletion of bmal1 was sufficient to trigger increased SeV susceptibility and correlated with impaired control of viral replication. Importantly, bmal1-/- mice developed much more extensive asthma-like airway changes post infection, including mucus production and increased airway resistance. In human airway samples from two asthma cohorts, we observed altered expression patterns of multiple clock genes. Our results suggest a role for bmal1 in the development of asthmatic airway disease via the regulation of lung antiviral responses to common viral triggers of asthma.
Collapse
Affiliation(s)
- Anna Ehlers
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Wenfang Xie
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Airport Road, Guangzhou, 510405, P.R. China
| | - Eugene Agapov
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Samuel Brown
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Deborah Steinberg
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Rose Tidwell
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Geneline Sajol
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Rebecca Schutz
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Rachel Weaver
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Huixi Yu
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Mario Castro
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Leonard B. Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Xinhua Wang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, 12 Airport Road, Guangzhou, 510405, P.R. China
| | - Michael J. Holtzman
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| | - Jeffrey A. Haspel
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO, 63110. USA
| |
Collapse
|
27
|
Stressed out: providing laboratory animals with behavioral control to reduce the physiological effects of stress. Lab Anim (NY) 2017; 46:142-145. [PMID: 28328902 DOI: 10.1038/laban.1218] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/13/2017] [Indexed: 12/31/2022]
Abstract
Laboratory animals experience a large amount of environmental stress. An animal's environment can include both physiological and social stressors that may require an animal to adapt to maintain allostatic balance. For example, thermal stress can lead to changes in behavior, reproduction and immune function, which has been detrimental to cancer modeling in mice. Chronic uncontrollable stress is widely acknowledged for its negative alterations to physiology. However, there is a lack in the understanding of how the laboratory environment affects animal physiology and behavior, particularly as it relates to characteristics of the human disease being modeled. Given the evidence on how stressors affect physiology, it is clear that efforts to model human physiology in animal models must consider animal stress as a confounding factor. We present evidence illustrating that providing captive animals with control or predictability is the best way to reduce the negative physiological effects of these difficult-to-manage stressors.
Collapse
|
28
|
Therapeutic dormancy to delay postsurgical glioma recurrence: the past, present and promise of focal hypothermia. J Neurooncol 2017; 133:447-454. [DOI: 10.1007/s11060-017-2471-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/07/2017] [Indexed: 01/06/2023]
|
29
|
Hylander BL, Eng JWL, Repasky EA. The Impact of Housing Temperature-Induced Chronic Stress on Preclinical Mouse Tumor Models and Therapeutic Responses: An Important Role for the Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:173-189. [PMID: 29275472 PMCID: PMC9423006 DOI: 10.1007/978-3-319-67577-0_12] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the last 10-15 years, there has been a recognition that the catecholamines (norepinephrine, NE, and epinephrine, Epi) released by the sympathetic nervous system under stressful conditions promote tumor growth through a variety of mechanisms. Tumors recruit autonomic nerves during their development and NE is then released locally in the tumor microenvironment (TME). Acting through adrenergic receptors present on a variety of cells in the TME, NE and Epi induce proliferation, resistance to apoptosis, epithelial to mesenchymal transition, metastasis of tumor cells, angiogenesis, and inflammation in the TME. These pre-clinical studies have been conducted in mouse models whose care and housing parameters are outlined in "The Guide for the Care and Use of Laboratory Animals [1]. In particular, the Guide mandates that mice be housed at standardized sub-thermoneutral temperatures; however, this causes a state of chronic cold-stress and elevated levels of NE. Although mice are able to maintain a normal body temperature when kept at these cool temperatures, it is becoming clear that this cold-stress is sufficient to activate physiological changes which affect experimental outcomes. We find that when mice are housed under standard, sub-thermoneutral temperatures (~22 °C, ST), tumor growth is significantly greater than when mice are housed at thermoneutrality (~30 °C TT). We also find that the anti-tumor immune response is suppressed at ST and this immunosuppression can be reversed by housing mice at TT or by administration of propranolol (a β-adrenergic receptor antagonist) to mice housed at ST. Furthermore, at ST tumors are more resistant to therapy and can also be sensitized to cytotoxic therapies by housing mice at TT or by treating mice with propranolol. The implications of these observations are particularly relevant to the way in which experiments conducted in preclinical models are interpreted and the findings implemented in the clinic. It may be that the disappointing failure of many new therapies to fulfill their promise in the clinic is related to an incomplete preclinical assessment in mouse models. Further, an expanded understanding of the efficacy of a therapy alone or in combination obtained by testing under a wider range of conditions would better predict how patients, who are under various levels of stress, might respond in a clinical setting. This may be particularly important to consider since we now appreciate that long term outcome of many therapies depends on eliciting an immune response.It is clear that the outcome of metabolic experiments, immunological investigations and therapeutic efficacy testing in tumors of mice housed at ST is restricted and expanding these experiments to include results obtained at TT may provide us with valuable information that would otherwise be overlooked.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jason W-L Eng
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
30
|
Seike I, Saito N, Saito S, Itoga M, Kayaba H. Influence of Average Income on Epidemics of Seasonal Influenza. Jpn J Infect Dis 2016; 69:457-463. [PMID: 26902220 DOI: 10.7883/yoken.jjid.2015.500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Understanding the local factors influencing the transmission of communicable diseases is important to minimize social damage. The aim of this study was to investigate local factors influencing seasonal influenza epidemics in Aomori prefecture consisting of 6 regions, i.e., Seihoku, Chunan, and Tosei on the west side, and Sanpachi, Kamikita, and Shimokita on the east side. Four indices (epidemic onset, duration, scale, and steepness of epidemic curves) were defined, and their correlations with regional characteristics and meteorological factors were investigated. Data for influenza seasons from 2006-2007 to 2014-2015 were collected. The 2009-2010 season was excluded because of the pandemic of A (H1N1)pdm09. Average income was strongly correlated with epidemic onset, duration, and scale. The ratio of children aged ≤5 years to the total population was strongly correlated with epidemic duration and scale. Low temperature in January showed moderate correlation with epidemic duration and scale. Cluster analysis showed that 2 isolated regions, Seihoku and Chunan, belonged to the same cluster in the 4 indices of epidemic curves, and other 2 relatively urbanized regions formed another cluster in 3 of the 4 indices. This study highlights important local factors that influence seasonal influenza epidemics and may help in implementation of preventive measures.
Collapse
|
31
|
Ma Y, Jia Y, Chen L, Ezeogu L, Yu B, Xu N, Liao DJ. Weaknesses and Pitfalls of Using Mice and Rats in Cancer Chemoprevention Studies. J Cancer 2015; 6:1058-65. [PMID: 26366220 PMCID: PMC4565856 DOI: 10.7150/jca.12519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/19/2015] [Indexed: 12/31/2022] Open
Abstract
Many studies, using different chemical agents, have shown excellent cancer prevention efficacy in mice and rats. However, equivalent tests of cancer prevention in humans require decades of intake of the agents while the rodents' short lifespans cannot give us information of the long-term safety. Therefore, animals with a much longer lifespan should be used to bridge the lifespan gap between the rodents and humans. There are many transgenic mouse models of carcinogenesis available, in which DNA promoters are used to activate transgenes. One promoter may activate the transgene in multiple cell types while different promoters are activated at different ages of the mice. These spatial and temporal aspects of transgenes are often neglected and may be pitfalls or weaknesses in chemoprevention studies. The variation in the copy number of the transgene may widen data variation and requires use of more animals. Models of chemically-induced carcinogenesis do not have these transgene-related defects, but chemical carcinogens usually damage metabolic organs or tissues, thus affecting the metabolism of the chemopreventive agents. Moreover, many genetically edited and some chemically-induced carcinogenesis models produce tumors that exhibit cancerous histology but are not cancers because the tumor cells are still mortal, inducer-dependent, and unable to metastasize, and thus should be used with caution in chemoprevention studies. Lastly, since mice prefer an ambient temperature of 30-32°C, it should be debated whether future mouse studies should be performed at this temperature, but not at 21-23°C that cold-stresses the animals.
Collapse
Affiliation(s)
- Yukui Ma
- 1. Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, P.R. China
| | - Yuping Jia
- 1. Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, P.R. China
| | - Lichan Chen
- 2. Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lewis Ezeogu
- 2. Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Baofa Yu
- 3. Beijing Baofa Cancer Hospital, Shahe Wangzhuang Gong Ye Yuan, Chang Pin Qu, Beijing 102206, P.R. China
| | - Ningzhi Xu
- 4. Laboratory of Cell and Molecular Biology, Cancer Institute, Chinese Academy of Medical Science, Beijing 100021, P.R. China
| | - D Joshua Liao
- 2. Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
32
|
Byrne KT, Vonderheide RH, Jaffee EM, Armstrong TD. Special Conference on Tumor Immunology and Immunotherapy: A New Chapter. Cancer Immunol Res 2015; 3:590-597. [PMID: 25968457 DOI: 10.1158/2326-6066.cir-15-0106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022]
Abstract
The overall objective of the fifth American Association for Cancer Research Special Conference, "Tumor Immunology and Immunotherapy: A New Chapter," organized by the Cancer Immunology Working Group, was to highlight multidisciplinary approaches of immunotherapy and mechanisms related to the ability of immunotherapy to fight established tumors. With the FDA approval of sipuleucel-T, ipilimumab (anti-CTLA-4; Bristol-Myers Squibb), and the two anti-PD-1 antibodies, pembrolizumab (formerly MK-3475 or lambrolizumab; Merck) and nivolumab (Bristol-Myers Squibb), immunotherapy has become a mainstream treatment option for some cancers. Many of the data presented at the conference and reviewed in this article showcase the progress made in determining the mechanistic reasons for the success of some treatments and the mechanisms associated with tolerance within the tumor microenvironment, both of which are potential targets for immunotherapy. In addition to combination and multimodal therapies, improvements in existing therapies will be needed to overcome the numerous ways that tumor-specific tolerance thwarts the immune system. This conference built upon the success of the 2012 conference and focused on seven progressing and/or emerging areas-new combination therapies, combination therapies and vaccine improvement, mechanisms of antibody therapy, factors in the tumor microenvironment affecting the immune response, the microbiomes effect on cancer and immunotherapy, metabolism in immunotherapy, and adoptive T-cell therapy. Cancer Immunol Res; 3(6); 1-8. ©2015 AACR.
Collapse
Affiliation(s)
- Katelyn T Byrne
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth M Jaffee
- Department of Oncology, Division of Gastrointestinal Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland. Skip Viragh Pancreatic Cancer Center, Johns Hopkins University, Baltimore, Maryland. Sol Goldman Pancreatic Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Todd D Armstrong
- Department of Oncology, Division of Gastrointestinal Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland. Skip Viragh Pancreatic Cancer Center, Johns Hopkins University, Baltimore, Maryland. Sol Goldman Pancreatic Cancer Center, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
33
|
Standard sub-thermoneutral caging temperature influences radiosensitivity of hematopoietic stem and progenitor cells. PLoS One 2015; 10:e0120078. [PMID: 25793392 PMCID: PMC4368554 DOI: 10.1371/journal.pone.0120078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/19/2015] [Indexed: 11/19/2022] Open
Abstract
The production of new blood cells relies on a hierarchical network of hematopoietic stem and progenitor cells (HSPCs). To maintain lifelong hematopoiesis, HSPCs must be protected from ionizing radiation or other cytotoxic agents. For many years, murine models have been a valuable source of information regarding factors that either enhance or reduce the survival of HSPCs after exposure of marrow to ionizing radiation. In a recent series of studies, however, it has become clear that housing-related factors such as the cool room temperature required for laboratory mice can exert a surprising influence on the outcome of experiments. Here we report that the mild, but chronic cold-stress endured by mice housed under these conditions exerts a protective effect on HSPCs after both non-lethal and lethal doses of total body irradiation (TBI). Alleviation of this cold-stress by housing mice at a thermoneutral temperature (30°C) resulted in significantly greater baseline radiosensitivity to a lethal dose of TBI with more HSPCs from mice housed at thermoneutral temperature undergoing apoptosis following non-lethal TBI. Cold-stressed mice have elevated levels of norepinephrine, a key molecule of the sympathetic nervous system that binds to β-adrenergic receptors. We show that blocking this signaling pathway in vivo through use of the β-blocker propanolol completely mitigates the protective effect of cold-stress on HSPC apoptosis. Collectively this study demonstrates that chronic stress endured by the standard housing conditions of laboratory mice increases the resistance of HSPCs to TBI-induced apoptosis through a mechanism that depends upon β-adrenergic signaling. Since β-blockers are commonly prescribed to a wide variety of patients, this information could be important when predicting the clinical impact of HSPC sensitivity to TBI.
Collapse
|