Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection

Ichiro Misumi, Joshua Starmer, Toru Uchimura, Melinda A Beck, Terry Magnuson, Jason K Whitmire, Ichiro Misumi, Joshua Starmer, Toru Uchimura, Melinda A Beck, Terry Magnuson, Jason K Whitmire

Abstract

Obesity in humans is associated with poorer health outcomes after infections compared with non-obese individuals. Here, we examined the effects of white adipose tissue and obesity on T cell responses to viral infection in mice. We show that lymphocytic choriomeningitis virus (LCMV) grows to high titer in adipose tissue. Virus-specific T cells enter the adipose tissue to resolve infection but then remain as a memory population distinct from memory T cells in lymphoid tissues. Memory T cells in adipose tissue are abundant in lean mice, and diet-induced obesity further increases memory T cell number in adipose tissue and spleen. Upon re-challenge infection, memory T cells rapidly cause severe pathogenesis, leading to increases in lipase levels, calcification of adipose tissue, pancreatitis, and reduced survival in obese mice but not lean mice. Thus, obesity leads to a unique form of viral pathogenesis involving memory T cell-dependent adipocyte destruction and damage to other tissues.

Keywords: LCMV; T cell memory; Trm; obesity; pancreatitis; tissue-resident memory T cells; white adipose tissue.

Conflict of interest statement

DECLARATION OF INTERESTS

The authors declare no competing interests.

Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. LCMV Infection of White Adipose…
Figure 1.. LCMV Infection of White Adipose Tissue
B6 mice were given LCMV-Armstrong (2 3 105 PFUs, intraperitoneal [i.p.]), and levels of virus in adipose and spleen and leukocyte abundance in adipose were determined. (A) The line graphs represent virus levels mean (±SEM) in the spleen or perigonadal white adipose tissue (WAT) at the indicated days after infection. The dotted horizontal line indicates the limit of detection. Data are pooled from two experiments with three to six mice per group. (B) Perigonadal fat pads were isolated from LCMV-infected mice at day 5 and assessed for viral infection. Gentle centrifugation was used to separate low density mature adipocytes from denser stromal vascular fraction (SVF) cell populations. The graph shows quantification of infectious virus (n = 3, one experiment), with an example of plaques developing in Vero cells. (C) Perigonadal adipose tissue was harvested at the indicated days of infection and analyzed by microscopy. Top: representative sections stained with H&E; scale bar, 120 mm. Bottom: whole-mount staining reveals the lipid droplet in adipocytes (BODIPYC12; red), cell nuclei (Hoechst; blue), and infiltrating T cells (anti-CD3; green); scale bar, 100 mm. Data are representative of two experiments with two or three mice per group. (D) Total number of leukocytes, CD8+ T cells, and CD4+ T cells recovered from fat pads before infection (n = 8) and at day 8 post-infection (n = 6). Data were analyzed using unpaired Student’s t test with **p

Figure 2.. Virus-Specific T Cells Accumulate in…

Figure 2.. Virus-Specific T Cells Accumulate in WAT

Virus-specific T cells in adipose tissue and…

Figure 2.. Virus-Specific T Cells Accumulate in WAT
Virus-specific T cells in adipose tissue and spleen were quantified at multiple times after acute infection by tetramer staining and ICCS. (A) Representative dot plots identify DbGP33+CD8+ T cells (left) and IAbGP67+CD4+ T cells (right) before infection and at days 8 and 70 post infection. The numbers indicate the frequency of the tetramer+ cells among all live cells in the spleen or among leukocytes isolated from the fat pads. (B) The number (mean ± SEM) of tetramer-positive T cells per spleen or fat pads at the indicated times post-infection. Data are drawn from two independent experiments with three to five mice per group. (C) Representative dot plots show GP33-specific CD8+ T cells or GP61-specific CD4+ T cells expression of IFNg after peptide stimulation. The analyses are from the indicated days after infection, with numbers indicating the percentage of IFNg+ cells among all cells. (D) The number (mean ± SEM) of IFNg+ epitopespecific T cells per tissue in spleen and fat pads as measured using ICCS assay at the indicated times post-infection. Data represent two experiments with five to nine mice per group. (E) T cell co-expression of IFNg with TNF or IFNg with IL-2 in the spleen or fat pads at day 8. CD8+ T cells were stimulated with GP33–41 peptide;CD4+ T cells were stimulated with GP61–80 peptide. See also Figure S1.

Figure 3.. A Distinct Subset of Memory…

Figure 3.. A Distinct Subset of Memory T Cells in WAT

(A and B) B6…

Figure 3.. A Distinct Subset of Memory T Cells in WAT
(A and B) B6 recipient mice (Ly5a–) were given TCR-transgenic (Tg+) 2 × 104 P14 CD8+ (Ly5a+) T cells or SMARTA CD4+ (Ly5a+) T cells and then infected with LCMV-Armstrong 1–2 days later. At day 38 post-infection, intravascular (i.v.) staining was performed to label circulating donor CD4+ or CD8+ T cells and distinguish them from T cells within the spleen or adipose tissue. (A) An illustration of the method whereby intravenous staining is coupled to ex vivo staining that labels all T cells in suspension. (B) Representative dot plots showing the proportion of T cells protected from intravenous staining because of their location within the parenchyma of the spleen or WAT. Note that the vast majority of T cells purified from WAT are not contaminants from circulating subsets. Data represent three independent experiments with three or four mice per group. (C and D) B6 recipient mice (Ly5a–) were given TCR-transgenic (Tg+) splenic 2 × 104 P14 (Ly5a) or SMARTA (Ly5a+) T cells and then infected with LCMV-Arm 1–2 days after engraftment. At day 38, spleen and WAT cells were isolated and co-stained to identify the donor T cells and their expression of phenotypic markers. (C) The histograms are gated on P14 memory cells (top) or SMARTA memory cells (bottom) and show their surface expression of the indicated molecules. Memory donor cells from spleen (red) or WAT (blue) are shown with naive splenic Tg+ cells (black). Data represent two experiments with four to six mice per group. (D) Memory P14 and SMARTA donor cells from day 50 post-infection were FACS-purified from the spleen or adipose tissue and subjected to RNA sequencing to identify changes in gene expression. The dot plots show donor cells from adipose tissue before and after FACS. The graphs show EdgeR analysis of the RNA-seq data with log fold change (logFC) in RNA expression in WAT T cells relative to splenic T cells plotted against theamount of RNA per cell, expressed as log counts per million reads (logCPM). The red circles identify significant (false discovery rate [FDR]

Figure 4.. Diet-Induced Obesity Increases Memory T…

Figure 4.. Diet-Induced Obesity Increases Memory T Cell Number

Mice were fed lean or high-fat…

Figure 4.. Diet-Induced Obesity Increases Memory T Cell Number
Mice were fed lean or high-fat diet for 8–9 weeks to establish obesity prior to LCMV-Armstrong infection. The number of virus-specific T cells in the spleen and fat pads was determined by tetramer staining and ICCS at multiple times after infection. (A) An illustration of the experimental approach. (B) The body weight of the mice and perigonadal fat pads at day 90 post-infection. (C) The line graphs show the number (mean ± SEM) of tetramer-positive T cells per spleen (top) or WAT (bottom) at the indicated times post infection. Data are pooled from two to four independent experiments with a total of three to nine mice per group. (D) The scatterplots show the number of GP33-specific or GP61-specific memory T cells that can make IFNg at day 90, as determined by ICCS assay. Each circle represents an individual mouse. Data represent four experiments with nine mice per group. (E) The line graphs represent the number (mean ± SEM) of DbGP33+CD8+ (left) or IAbGP67+CD4+ (right) T cells per WAT divided by the weight of the fat pad. Data represent two or four experiments with three to nine mice per group. (F) Lymphocytes were isolated from the spleen and fat pads of lean or obese mice at day 90 post infection and exposed to the indicated peptide in an ICCS assay. The graphs show the geometric mean fluorescence intensity (+SEM) among IFNg expression by T cells. Data represent four experiments with nine mice per group. Significance determined by unpaired Student’s t test: *p

Figure 5.. Immune Obese Mice Develop T…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge

Cohorts of…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge
Cohorts of mice were fed lean or high-fat diet starting at 4–5 weeks of age until 13–15 weeks of age. Lean mice were 40 g (n = 13). Some of the mice were immunized with LCMV-Armstrong, and the rest were left LCMV naive. Sixty-five days later, the mice were challenged with LCMV-Clone13 (2 × 106 PFUs, i.p.) and analyzed 2 or 5 days later. (A) An illustration of the experimental approach. (B) Survival of naive (solid lines) and immune (dashed lines) mice after challenge, including mice requiring humane euthanasia. Among the re-challenged LCMV-immune mice, a Mantel-Cox log rank test revealed a significant difference (p = 0.0039) between the naive and obese mice. Data represent 7–16 mice/group. (C) Lipase activity in ascites (left) and serum (right) at 2 days post-challenge infection. Dots on figure represent individual mice (3–6 per group, two independent experiments). (D) Representative pancreas sections from the indicated four groups of mice at 2 days post-challenge infection. Sections were stained with H&E (upper two rows) or Von Kossa stain to reveal calcium deposition (dark gray, bottom row). The upper images show regions within the pancreas; the middle bottom images show the margin of the pancreas. Scale bar, 150 μm. (E) Concentrations of IL-6, IFNg, and TNF in blood. Symbols represent individual mice (3 or 4 per group, two independent experiments). Significance determined by unpaired Student’s t test: *p

Figure 6.. Memory T Cells Mediate Acute…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis

Cohorts of lean…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis
Cohorts of lean or obese immune mice were treated with antibodies to CD8 and CD4 to deplete T cells or treated with isotype control antibodies before challenge. Mice were analyzed 2 days after infection. Data compiled from two independent experiments with four to six mice per group. (A) Lipase activity in the peritoneal cavity. (B) Sections of pancreas and adipose stained with Von Kossa. Scale bar, 150 μm. (C) Calcium concentrations in blood. (D) ALT concentration in blood. (E) Body temperature in individual mice. Significance determined by unpaired Student’s t test: *p
Similar articles
Cited by
References
    1. Ahmed A, Azim A, Gurjar M, and Baronia AK (2016). Hypocalcemia in acute pancreatitis revisited. Indian J. Crit. Care Med 20, 173–177. - PMC - PubMed
    1. Anderson KG, Mayer-Barber K, Sung H, Beura L, James BR, Taylor JJ, Qunaj L, Griffith TS, Vezys V, Barber DL, and Masopust D (2014). Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc 9, 209–222. - PMC - PubMed
    1. Barnes MA, Carson MJ, and Nair MG (2015). Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine 72, 210–219. - PMC - PubMed
    1. Chen AT, Cornberg M, Gras S, Guillonneau C, Rossjohn J, Trees A, Emonet S, de la Torre JC, Welsh RM, and Selin LK (2012). Loss of anti-viral immunity by infection with a virus encoding a cross-reactive pathogenic epitope. PLoS Pathog 8, e1002633. - PMC - PubMed
    1. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, and Obin MS (2005). Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res 46, 2347–2355. - PubMed
Show all 41 references
Publication types
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2.. Virus-Specific T Cells Accumulate in…
Figure 2.. Virus-Specific T Cells Accumulate in WAT
Virus-specific T cells in adipose tissue and spleen were quantified at multiple times after acute infection by tetramer staining and ICCS. (A) Representative dot plots identify DbGP33+CD8+ T cells (left) and IAbGP67+CD4+ T cells (right) before infection and at days 8 and 70 post infection. The numbers indicate the frequency of the tetramer+ cells among all live cells in the spleen or among leukocytes isolated from the fat pads. (B) The number (mean ± SEM) of tetramer-positive T cells per spleen or fat pads at the indicated times post-infection. Data are drawn from two independent experiments with three to five mice per group. (C) Representative dot plots show GP33-specific CD8+ T cells or GP61-specific CD4+ T cells expression of IFNg after peptide stimulation. The analyses are from the indicated days after infection, with numbers indicating the percentage of IFNg+ cells among all cells. (D) The number (mean ± SEM) of IFNg+ epitopespecific T cells per tissue in spleen and fat pads as measured using ICCS assay at the indicated times post-infection. Data represent two experiments with five to nine mice per group. (E) T cell co-expression of IFNg with TNF or IFNg with IL-2 in the spleen or fat pads at day 8. CD8+ T cells were stimulated with GP33–41 peptide;CD4+ T cells were stimulated with GP61–80 peptide. See also Figure S1.
Figure 3.. A Distinct Subset of Memory…
Figure 3.. A Distinct Subset of Memory T Cells in WAT
(A and B) B6 recipient mice (Ly5a–) were given TCR-transgenic (Tg+) 2 × 104 P14 CD8+ (Ly5a+) T cells or SMARTA CD4+ (Ly5a+) T cells and then infected with LCMV-Armstrong 1–2 days later. At day 38 post-infection, intravascular (i.v.) staining was performed to label circulating donor CD4+ or CD8+ T cells and distinguish them from T cells within the spleen or adipose tissue. (A) An illustration of the method whereby intravenous staining is coupled to ex vivo staining that labels all T cells in suspension. (B) Representative dot plots showing the proportion of T cells protected from intravenous staining because of their location within the parenchyma of the spleen or WAT. Note that the vast majority of T cells purified from WAT are not contaminants from circulating subsets. Data represent three independent experiments with three or four mice per group. (C and D) B6 recipient mice (Ly5a–) were given TCR-transgenic (Tg+) splenic 2 × 104 P14 (Ly5a) or SMARTA (Ly5a+) T cells and then infected with LCMV-Arm 1–2 days after engraftment. At day 38, spleen and WAT cells were isolated and co-stained to identify the donor T cells and their expression of phenotypic markers. (C) The histograms are gated on P14 memory cells (top) or SMARTA memory cells (bottom) and show their surface expression of the indicated molecules. Memory donor cells from spleen (red) or WAT (blue) are shown with naive splenic Tg+ cells (black). Data represent two experiments with four to six mice per group. (D) Memory P14 and SMARTA donor cells from day 50 post-infection were FACS-purified from the spleen or adipose tissue and subjected to RNA sequencing to identify changes in gene expression. The dot plots show donor cells from adipose tissue before and after FACS. The graphs show EdgeR analysis of the RNA-seq data with log fold change (logFC) in RNA expression in WAT T cells relative to splenic T cells plotted against theamount of RNA per cell, expressed as log counts per million reads (logCPM). The red circles identify significant (false discovery rate [FDR]

Figure 4.. Diet-Induced Obesity Increases Memory T…

Figure 4.. Diet-Induced Obesity Increases Memory T Cell Number

Mice were fed lean or high-fat…

Figure 4.. Diet-Induced Obesity Increases Memory T Cell Number
Mice were fed lean or high-fat diet for 8–9 weeks to establish obesity prior to LCMV-Armstrong infection. The number of virus-specific T cells in the spleen and fat pads was determined by tetramer staining and ICCS at multiple times after infection. (A) An illustration of the experimental approach. (B) The body weight of the mice and perigonadal fat pads at day 90 post-infection. (C) The line graphs show the number (mean ± SEM) of tetramer-positive T cells per spleen (top) or WAT (bottom) at the indicated times post infection. Data are pooled from two to four independent experiments with a total of three to nine mice per group. (D) The scatterplots show the number of GP33-specific or GP61-specific memory T cells that can make IFNg at day 90, as determined by ICCS assay. Each circle represents an individual mouse. Data represent four experiments with nine mice per group. (E) The line graphs represent the number (mean ± SEM) of DbGP33+CD8+ (left) or IAbGP67+CD4+ (right) T cells per WAT divided by the weight of the fat pad. Data represent two or four experiments with three to nine mice per group. (F) Lymphocytes were isolated from the spleen and fat pads of lean or obese mice at day 90 post infection and exposed to the indicated peptide in an ICCS assay. The graphs show the geometric mean fluorescence intensity (+SEM) among IFNg expression by T cells. Data represent four experiments with nine mice per group. Significance determined by unpaired Student’s t test: *p

Figure 5.. Immune Obese Mice Develop T…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge

Cohorts of…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge
Cohorts of mice were fed lean or high-fat diet starting at 4–5 weeks of age until 13–15 weeks of age. Lean mice were 40 g (n = 13). Some of the mice were immunized with LCMV-Armstrong, and the rest were left LCMV naive. Sixty-five days later, the mice were challenged with LCMV-Clone13 (2 × 106 PFUs, i.p.) and analyzed 2 or 5 days later. (A) An illustration of the experimental approach. (B) Survival of naive (solid lines) and immune (dashed lines) mice after challenge, including mice requiring humane euthanasia. Among the re-challenged LCMV-immune mice, a Mantel-Cox log rank test revealed a significant difference (p = 0.0039) between the naive and obese mice. Data represent 7–16 mice/group. (C) Lipase activity in ascites (left) and serum (right) at 2 days post-challenge infection. Dots on figure represent individual mice (3–6 per group, two independent experiments). (D) Representative pancreas sections from the indicated four groups of mice at 2 days post-challenge infection. Sections were stained with H&E (upper two rows) or Von Kossa stain to reveal calcium deposition (dark gray, bottom row). The upper images show regions within the pancreas; the middle bottom images show the margin of the pancreas. Scale bar, 150 μm. (E) Concentrations of IL-6, IFNg, and TNF in blood. Symbols represent individual mice (3 or 4 per group, two independent experiments). Significance determined by unpaired Student’s t test: *p

Figure 6.. Memory T Cells Mediate Acute…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis

Cohorts of lean…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis
Cohorts of lean or obese immune mice were treated with antibodies to CD8 and CD4 to deplete T cells or treated with isotype control antibodies before challenge. Mice were analyzed 2 days after infection. Data compiled from two independent experiments with four to six mice per group. (A) Lipase activity in the peritoneal cavity. (B) Sections of pancreas and adipose stained with Von Kossa. Scale bar, 150 μm. (C) Calcium concentrations in blood. (D) ALT concentration in blood. (E) Body temperature in individual mice. Significance determined by unpaired Student’s t test: *p
Similar articles
Cited by
References
    1. Ahmed A, Azim A, Gurjar M, and Baronia AK (2016). Hypocalcemia in acute pancreatitis revisited. Indian J. Crit. Care Med 20, 173–177. - PMC - PubMed
    1. Anderson KG, Mayer-Barber K, Sung H, Beura L, James BR, Taylor JJ, Qunaj L, Griffith TS, Vezys V, Barber DL, and Masopust D (2014). Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc 9, 209–222. - PMC - PubMed
    1. Barnes MA, Carson MJ, and Nair MG (2015). Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine 72, 210–219. - PMC - PubMed
    1. Chen AT, Cornberg M, Gras S, Guillonneau C, Rossjohn J, Trees A, Emonet S, de la Torre JC, Welsh RM, and Selin LK (2012). Loss of anti-viral immunity by infection with a virus encoding a cross-reactive pathogenic epitope. PLoS Pathog 8, e1002633. - PMC - PubMed
    1. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, and Obin MS (2005). Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res 46, 2347–2355. - PubMed
Show all 41 references
Publication types
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4.. Diet-Induced Obesity Increases Memory T…
Figure 4.. Diet-Induced Obesity Increases Memory T Cell Number
Mice were fed lean or high-fat diet for 8–9 weeks to establish obesity prior to LCMV-Armstrong infection. The number of virus-specific T cells in the spleen and fat pads was determined by tetramer staining and ICCS at multiple times after infection. (A) An illustration of the experimental approach. (B) The body weight of the mice and perigonadal fat pads at day 90 post-infection. (C) The line graphs show the number (mean ± SEM) of tetramer-positive T cells per spleen (top) or WAT (bottom) at the indicated times post infection. Data are pooled from two to four independent experiments with a total of three to nine mice per group. (D) The scatterplots show the number of GP33-specific or GP61-specific memory T cells that can make IFNg at day 90, as determined by ICCS assay. Each circle represents an individual mouse. Data represent four experiments with nine mice per group. (E) The line graphs represent the number (mean ± SEM) of DbGP33+CD8+ (left) or IAbGP67+CD4+ (right) T cells per WAT divided by the weight of the fat pad. Data represent two or four experiments with three to nine mice per group. (F) Lymphocytes were isolated from the spleen and fat pads of lean or obese mice at day 90 post infection and exposed to the indicated peptide in an ICCS assay. The graphs show the geometric mean fluorescence intensity (+SEM) among IFNg expression by T cells. Data represent four experiments with nine mice per group. Significance determined by unpaired Student’s t test: *p

Figure 5.. Immune Obese Mice Develop T…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge

Cohorts of…

Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge
Cohorts of mice were fed lean or high-fat diet starting at 4–5 weeks of age until 13–15 weeks of age. Lean mice were 40 g (n = 13). Some of the mice were immunized with LCMV-Armstrong, and the rest were left LCMV naive. Sixty-five days later, the mice were challenged with LCMV-Clone13 (2 × 106 PFUs, i.p.) and analyzed 2 or 5 days later. (A) An illustration of the experimental approach. (B) Survival of naive (solid lines) and immune (dashed lines) mice after challenge, including mice requiring humane euthanasia. Among the re-challenged LCMV-immune mice, a Mantel-Cox log rank test revealed a significant difference (p = 0.0039) between the naive and obese mice. Data represent 7–16 mice/group. (C) Lipase activity in ascites (left) and serum (right) at 2 days post-challenge infection. Dots on figure represent individual mice (3–6 per group, two independent experiments). (D) Representative pancreas sections from the indicated four groups of mice at 2 days post-challenge infection. Sections were stained with H&E (upper two rows) or Von Kossa stain to reveal calcium deposition (dark gray, bottom row). The upper images show regions within the pancreas; the middle bottom images show the margin of the pancreas. Scale bar, 150 μm. (E) Concentrations of IL-6, IFNg, and TNF in blood. Symbols represent individual mice (3 or 4 per group, two independent experiments). Significance determined by unpaired Student’s t test: *p

Figure 6.. Memory T Cells Mediate Acute…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis

Cohorts of lean…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis
Cohorts of lean or obese immune mice were treated with antibodies to CD8 and CD4 to deplete T cells or treated with isotype control antibodies before challenge. Mice were analyzed 2 days after infection. Data compiled from two independent experiments with four to six mice per group. (A) Lipase activity in the peritoneal cavity. (B) Sections of pancreas and adipose stained with Von Kossa. Scale bar, 150 μm. (C) Calcium concentrations in blood. (D) ALT concentration in blood. (E) Body temperature in individual mice. Significance determined by unpaired Student’s t test: *p
Similar articles
Cited by
References
    1. Ahmed A, Azim A, Gurjar M, and Baronia AK (2016). Hypocalcemia in acute pancreatitis revisited. Indian J. Crit. Care Med 20, 173–177. - PMC - PubMed
    1. Anderson KG, Mayer-Barber K, Sung H, Beura L, James BR, Taylor JJ, Qunaj L, Griffith TS, Vezys V, Barber DL, and Masopust D (2014). Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc 9, 209–222. - PMC - PubMed
    1. Barnes MA, Carson MJ, and Nair MG (2015). Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine 72, 210–219. - PMC - PubMed
    1. Chen AT, Cornberg M, Gras S, Guillonneau C, Rossjohn J, Trees A, Emonet S, de la Torre JC, Welsh RM, and Selin LK (2012). Loss of anti-viral immunity by infection with a virus encoding a cross-reactive pathogenic epitope. PLoS Pathog 8, e1002633. - PMC - PubMed
    1. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, and Obin MS (2005). Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res 46, 2347–2355. - PubMed
Show all 41 references
Publication types
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5.. Immune Obese Mice Develop T…
Figure 5.. Immune Obese Mice Develop T Cell-Dependent Lethal Acute Pancreatitis upon Re-challenge
Cohorts of mice were fed lean or high-fat diet starting at 4–5 weeks of age until 13–15 weeks of age. Lean mice were 40 g (n = 13). Some of the mice were immunized with LCMV-Armstrong, and the rest were left LCMV naive. Sixty-five days later, the mice were challenged with LCMV-Clone13 (2 × 106 PFUs, i.p.) and analyzed 2 or 5 days later. (A) An illustration of the experimental approach. (B) Survival of naive (solid lines) and immune (dashed lines) mice after challenge, including mice requiring humane euthanasia. Among the re-challenged LCMV-immune mice, a Mantel-Cox log rank test revealed a significant difference (p = 0.0039) between the naive and obese mice. Data represent 7–16 mice/group. (C) Lipase activity in ascites (left) and serum (right) at 2 days post-challenge infection. Dots on figure represent individual mice (3–6 per group, two independent experiments). (D) Representative pancreas sections from the indicated four groups of mice at 2 days post-challenge infection. Sections were stained with H&E (upper two rows) or Von Kossa stain to reveal calcium deposition (dark gray, bottom row). The upper images show regions within the pancreas; the middle bottom images show the margin of the pancreas. Scale bar, 150 μm. (E) Concentrations of IL-6, IFNg, and TNF in blood. Symbols represent individual mice (3 or 4 per group, two independent experiments). Significance determined by unpaired Student’s t test: *p

Figure 6.. Memory T Cells Mediate Acute…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis

Cohorts of lean…

Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis
Cohorts of lean or obese immune mice were treated with antibodies to CD8 and CD4 to deplete T cells or treated with isotype control antibodies before challenge. Mice were analyzed 2 days after infection. Data compiled from two independent experiments with four to six mice per group. (A) Lipase activity in the peritoneal cavity. (B) Sections of pancreas and adipose stained with Von Kossa. Scale bar, 150 μm. (C) Calcium concentrations in blood. (D) ALT concentration in blood. (E) Body temperature in individual mice. Significance determined by unpaired Student’s t test: *p
Similar articles
Cited by
References
    1. Ahmed A, Azim A, Gurjar M, and Baronia AK (2016). Hypocalcemia in acute pancreatitis revisited. Indian J. Crit. Care Med 20, 173–177. - PMC - PubMed
    1. Anderson KG, Mayer-Barber K, Sung H, Beura L, James BR, Taylor JJ, Qunaj L, Griffith TS, Vezys V, Barber DL, and Masopust D (2014). Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc 9, 209–222. - PMC - PubMed
    1. Barnes MA, Carson MJ, and Nair MG (2015). Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine 72, 210–219. - PMC - PubMed
    1. Chen AT, Cornberg M, Gras S, Guillonneau C, Rossjohn J, Trees A, Emonet S, de la Torre JC, Welsh RM, and Selin LK (2012). Loss of anti-viral immunity by infection with a virus encoding a cross-reactive pathogenic epitope. PLoS Pathog 8, e1002633. - PMC - PubMed
    1. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, and Obin MS (2005). Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res 46, 2347–2355. - PubMed
Show all 41 references
Publication types
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6.. Memory T Cells Mediate Acute…
Figure 6.. Memory T Cells Mediate Acute Pancreatitis and Adipose Tissue Necrosis
Cohorts of lean or obese immune mice were treated with antibodies to CD8 and CD4 to deplete T cells or treated with isotype control antibodies before challenge. Mice were analyzed 2 days after infection. Data compiled from two independent experiments with four to six mice per group. (A) Lipase activity in the peritoneal cavity. (B) Sections of pancreas and adipose stained with Von Kossa. Scale bar, 150 μm. (C) Calcium concentrations in blood. (D) ALT concentration in blood. (E) Body temperature in individual mice. Significance determined by unpaired Student’s t test: *p

References

    1. Ahmed A, Azim A, Gurjar M, and Baronia AK (2016). Hypocalcemia in acute pancreatitis revisited. Indian J. Crit. Care Med 20, 173–177.
    1. Anderson KG, Mayer-Barber K, Sung H, Beura L, James BR, Taylor JJ, Qunaj L, Griffith TS, Vezys V, Barber DL, and Masopust D (2014). Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc 9, 209–222.
    1. Barnes MA, Carson MJ, and Nair MG (2015). Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine 72, 210–219.
    1. Chen AT, Cornberg M, Gras S, Guillonneau C, Rossjohn J, Trees A, Emonet S, de la Torre JC, Welsh RM, and Selin LK (2012). Loss of anti-viral immunity by infection with a virus encoding a cross-reactive pathogenic epitope. PLoS Pathog 8, e1002633.
    1. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, and Obin MS (2005). Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res 46, 2347–2355.
    1. Couturier J, Suliburk JW, Brown JM, Luke DJ, Agarwal N, Yu X, Nguyen C, Iyer D, Kozinetz CA, Overbeek PA, et al. (2015). Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. AIDS 29, 667–674.
    1. Deng T, Lyon CJ, Minze LJ, Lin J, Zou J, Liu JZ, Ren Y, Yin Z, Hamilton DJ, Reardon PR, et al. (2013). Class II major histocompatibility complex plays an essential role in obesity-induced adipose inflammation. Cell Metab 17, 411–422.
    1. Dettelbach MA, Deftos LJ, and Stewart AF (1990). Intraperitoneal free fatty acids induce severe hypocalcemia in rats: a model for the hypocalcemia of pancreatitis. J. Bone Miner. Res 5, 1249–1255.
    1. Díaz E, Rodríguez A, Martin-Loeches I, Lorente L, Del Mar Martín M, Pozo JC, Montejo JC, Estella A, Arenzana Á , and Rello J; H1N1 SEMICYUC Working Group (2011). Impact of obesity in patients infected with 2009 influenza A(H1N1). Chest 139, 382–386.
    1. Gerlach C, Moseman EA, Loughhead SM, Alvarez D, Zwijnenburg AJ, Waanders L, Garg R, de la Torre JC, and von Andrian UH (2016). The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 45, 1270–1284.
    1. Grant RW, and Dixit VD (2015). Adipose tissue as an immunological organ. Obesity (Silver Spring) 23, 512–518.
    1. Grant R, Youm YH, Ravussin A, and Dixit VD (2013). Quantification of adipose tissue leukocytosis in obesity. Methods Mol. Biol 1040, 195–209.
    1. Han SJ, Glatman Zaretsky A, Andrade-Oliveira V, Collins N, Dzutsev A, Shaik J, Morais da Fonseca D, Harrison OJ, Tamoutounour S, Byrd AL, et al. (2017). White adipose tissue is a reservoir for memory T cells and promotes protective memory responses to infection. Immunity 47, 1154–1168.e6.
    1. Herndler-Brandstetter D, Ishigame H, Shinnakasu R, Plajer V, Stecher C, Zhao J, Lietzenmayer M, Kroehling L, Takumi A, Kometani K, et al. (2018). KLRG1(+) effector CD8(+) T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective immunity. Immunity 48, 716–729.e8.
    1. Huh JY, Park YJ, Ham M, and Kim JB (2014). Crosstalk between adipocytes and immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. Mol. Cells 37, 365–371.
    1. Huttunen R, and Syrjänen J (2013). Obesity and the risk and outcome of infection. Int. J. Obes 37, 333–340.
    1. Karlsson EA, Sheridan PA, and Beck MA (2010). Diet-induced obesity impairs the T cell memory response to influenza virus infection. J. Immunol 184, 3127–3133.
    1. Malide D (2008). Application of immunocytochemistry and immunofluorescence techniques to adipose tissue and cell cultures. Methods Mol. Biol 456, 285–297.
    1. Marshall HD, Chandele A, Jung YW, Meng H, Poholek AC, Parish IA, Rutishauser R, Cui W, Kleinstein SH, Craft J, and Kaech SM (2011). Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4(+) cell properties during viral infection. Immunity 35, 633–646.
    1. Martínez J, Johnson CD, Sánchez-Payá J, de Madaria E, Robles-Díaz G, and Pérez-Mateo M (2006). Obesity is a definitive risk factor of severity and mortality in acute pancreatitis: an updated meta-analysis. Pancreatology 6, 206–209.
    1. Masopust D, Murali-Krishna K, and Ahmed R (2007). Quantitating the magnitude of the lymphocytic choriomeningitis virus-specific CD8 T-cell response: it is even bigger than we thought. J. Virol 81, 2002–2011.
    1. Milner JJ, and Beck MA (2012). The impact of obesity on the immune response to infection. Proc. Nutr. Soc 71, 298–306.
    1. Moraes-Vieira PM, Larocca RA, Bassi EJ, Peron JP, Andrade-Oliveira V, Wasinski F, Araujo R, Thornley T, Quintana FJ, Basso AS, et al. (2014). Leptin deficiency impairs maturation of dendritic cells and enhances induction of regulatory T and Th17 cells. Eur. J. Immunol 44, 794–806.
    1. Navina S, Acharya C, DeLany JP, Orlichenko LS, Baty CJ, Shiva SS, Durgampudi C, Karlsson JM, Lee K, Bae KT, et al. (2011). Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci. Transl. Med 3, 107ra110.
    1. Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K, Sugiura S, et al. (2009). CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med 15, 914–920.
    1. O’Brien KB, Vogel P, Duan S, Govorkova EA, Webby RJ, McCullers JA, and Schultz-Cherry S (2012). Impaired wound healing predisposes obese mice to severe influenza virus infection. J. Infect. Dis 205, 252–261.
    1. Papathanassoglou E, El-Haschimi K, Li XC, Matarese G, Strom T, and Mantzoros C (2006). Leptin receptor expression and signaling in lymphocytes: kinetics during lymphocyte activation, role in lymphocyte survival, and response to high fat diet in mice. J. Immunol 176, 7745–7752.
    1. Park J, Chang JH, Park SH, Lee HJ, Lim YS, Kim TH, Kim CW, and Han SW (2015). Interleukin-6 is associated with obesity, central fat distribution, and disease severity in patients with acute pancreatitis. Pancreatology 15, 59–63.
    1. Patel K, Trivedi RN, Durgampudi C, Noel P, Cline RA, DeLany JP, Navina S, and Singh VP (2015). Lipolysis of visceral adipocyte triglyceride by pancreatic lipases converts mild acute pancreatitis to severe pancreatitis independent of necrosis and inflammation. Am. J. Pathol 185, 808–819.
    1. Procaccini C, La Rocca C, Carbone F, De Rosa V, Galgani M, and Matarese G (2017). Leptin as immune mediator: Interaction between neuroendocrine and immune system. Dev. Comp. Immunol 66, 120–129.
    1. Rosen ED, and Spiegelman BM (2014). What we talk about when we talk about fat. Cell 156, 20–44.
    1. Schenkel JM, and Masopust D (2014). Tissue-resident memory T cells. Immunity 41, 886–897.
    1. Selin LK, Varga SM, Wong IC, and Welsh RM (1998). Protective heterologous antiviral immunity and enhanced immunopathogenesis mediated by memory T cell populations. J. Exp. Med 188, 1705–1715.
    1. Sheridan PA, Paich HA, Handy J, Karlsson EA, Hudgens MG, Sammon AB, Holland LA, Weir S, Noah TL, and Beck MA (2012). Obesity is associated with impaired immune response to influenza vaccination in humans. Int. J. Obes 36, 1072–1077.
    1. Shin H, and Iwasaki A (2013). Tissue-resident memory T cells. Immunol. Rev 255, 165–181.
    1. Smith AG, Sheridan PA, Harp JB, and Beck MA (2007). Diet-induced obese mice have increased mortality and altered immune responses when infected with influenza virus. J. Nutr 137, 1236–1243.
    1. Teixeira L, Marques RM, Ferreirinha P, Bezerra F, Melo J, Moreira J, Pinto A, Correia A, Ferreira PG, and Vilanova M (2016). Enrichment of IFN-g producing cells in different murine adipose tissue depots upon infection with an apicomplexan parasite. Sci. Rep 6, 23475.
    1. Tsatsanis C, Margioris AN, and Kontoyiannis DP (2010). Association between H1N1 infection severity and obesity-adiponectin as a potential etiologic factor. J. Infect. Dis 202, 459–460.
    1. Twig G, Geva N, Levine H, Derazne E, Goldberger N, Haklai Z, Leiba A, and Kark JD (2018). Body mass index and infectious disease mortality in midlife in a cohort of 2.3 million adolescents. Int. J. Obes. (Lond.) 42, 801–807.
    1. Welsh RM, Che JW, Brehm MA, and Selin LK (2010). Heterologous immunity between viruses. Immunol. Rev 235, 244–266.
    1. Yang HY, Joris I, Majno G, and Welsh RM (1985). Necrosis of adipose tissue induced by sequential infections with unrelated viruses. Am. J. Pathol 120, 173–177.

Source: PubMed

3
구독하다