author_facet Miller, Janice
Dreczkowski, Gillian
Ramage, Michael I.
Wigmore, Stephen J.
Gallagher, Iain J.
Skipworth, Richard J.E.
Miller, Janice
Dreczkowski, Gillian
Ramage, Michael I.
Wigmore, Stephen J.
Gallagher, Iain J.
Skipworth, Richard J.E.
author Miller, Janice
Dreczkowski, Gillian
Ramage, Michael I.
Wigmore, Stephen J.
Gallagher, Iain J.
Skipworth, Richard J.E.
spellingShingle Miller, Janice
Dreczkowski, Gillian
Ramage, Michael I.
Wigmore, Stephen J.
Gallagher, Iain J.
Skipworth, Richard J.E.
Journal of Cachexia, Sarcopenia and Muscle
Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
Physiology (medical)
Orthopedics and Sports Medicine
author_sort miller, janice
spelling Miller, Janice Dreczkowski, Gillian Ramage, Michael I. Wigmore, Stephen J. Gallagher, Iain J. Skipworth, Richard J.E. 2190-5991 2190-6009 Wiley Physiology (medical) Orthopedics and Sports Medicine http://dx.doi.org/10.1002/jcsm.12568 <jats:title>Abstract</jats:title><jats:sec><jats:title>Background</jats:title><jats:p>Cancer cachexia is a poorly understood metabolic consequence of cancer. During cachexia, different adipose depots demonstrate differential wasting rates. Animal models suggest adipose tissue may be a key driver of muscle wasting through fat–muscle crosstalk, but human studies in this area are lacking. We performed global gene expression profiling of visceral (VAT) and subcutaneous (SAT) adipose from weight stable and cachectic cancer patients and healthy controls.</jats:p></jats:sec><jats:sec><jats:title>Methods</jats:title><jats:p>Cachexia was defined as &gt;2% weight loss plus low computed tomography‐muscularity. Biopsies of SAT and VAT were taken from patients undergoing resection for oesophago‐gastric cancer, and healthy controls (<jats:italic>n</jats:italic> = 16 and 8 respectively). RNA was isolated and reverse transcribed. cDNA was hybridised to the Affymetrix Clariom S microarray and data analysed using R/Bioconductor. Differential expression of genes was assessed using empirical Bayes and moderated‐<jats:italic>t</jats:italic>‐statistic approaches. Category enrichment analysis was used with a tissue‐specific background to examine the biological context of differentially expressed genes. Selected differentially regulated genes were validated by qPCR. Enzyme‐linked immunosorbent assay (ELISA) for intelectin‐1 was performed on all VAT samples. The previously‐described cohort plus 12 additional patients from each group also had plasma I = intelectin‐1 ELISA carried out.</jats:p></jats:sec><jats:sec><jats:title>Results</jats:title><jats:p>In VAT vs. SAT comparisons, there were 2101, 1722, and 1659 significantly regulated genes in the cachectic, weight stable, and control groups, respectively. There were 2200 significantly regulated genes from VAT in cachectic patients compared with controls. Genes involving inflammation were enriched in cancer and control VAT vs. SAT, although different genes contributed to enrichment in each group. Energy metabolism, fat browning (e.g. uncoupling protein 1), and adipogenesis genes were down‐regulated in cancer VAT <jats:italic>(P</jats:italic> = 0.043, <jats:italic>P</jats:italic> = 5.4 × 10<jats:sup>−6</jats:sup> and <jats:italic>P</jats:italic> = 1 × 10<jats:sup>−6</jats:sup> respectively). The gene showing the largest difference in expression was ITLN1, the gene that encodes for intelectin‐1 (false discovery rate‐corrected <jats:italic>P</jats:italic> = 0.0001), a novel adipocytokine associated with weight loss in other contexts.</jats:p></jats:sec><jats:sec><jats:title>Conclusions</jats:title><jats:p>SAT and VAT have unique gene expression signatures in cancer and cachexia. VAT is metabolically active in cancer, and intelectin‐1 may be a target for therapeutic manipulation. VAT may play a fundamental role in cachexia, but the down‐regulation of energy metabolism genes implies a limited role for fat browning in cachectic patients, in contrast to pre‐clinical models.</jats:p></jats:sec> Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia Journal of Cachexia, Sarcopenia and Muscle
doi_str_mv 10.1002/jcsm.12568
facet_avail Online
Free
finc_class_facet Biologie
Medizin
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi9qY3NtLjEyNTY4
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi9qY3NtLjEyNTY4
institution DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
imprint Wiley, 2020
imprint_str_mv Wiley, 2020
issn 2190-5991
2190-6009
issn_str_mv 2190-5991
2190-6009
language English
mega_collection Wiley (CrossRef)
match_str miller2020adiposedepotgeneexpressionandintelectin1inthemetabolicresponsetocancerandcachexia
publishDateSort 2020
publisher Wiley
recordtype ai
record_format ai
series Journal of Cachexia, Sarcopenia and Muscle
source_id 49
title Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_unstemmed Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_full Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_fullStr Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_full_unstemmed Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_short Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_sort adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
topic Physiology (medical)
Orthopedics and Sports Medicine
url http://dx.doi.org/10.1002/jcsm.12568
publishDate 2020
physical 1141-1153
description <jats:title>Abstract</jats:title><jats:sec><jats:title>Background</jats:title><jats:p>Cancer cachexia is a poorly understood metabolic consequence of cancer. During cachexia, different adipose depots demonstrate differential wasting rates. Animal models suggest adipose tissue may be a key driver of muscle wasting through fat–muscle crosstalk, but human studies in this area are lacking. We performed global gene expression profiling of visceral (VAT) and subcutaneous (SAT) adipose from weight stable and cachectic cancer patients and healthy controls.</jats:p></jats:sec><jats:sec><jats:title>Methods</jats:title><jats:p>Cachexia was defined as &gt;2% weight loss plus low computed tomography‐muscularity. Biopsies of SAT and VAT were taken from patients undergoing resection for oesophago‐gastric cancer, and healthy controls (<jats:italic>n</jats:italic> = 16 and 8 respectively). RNA was isolated and reverse transcribed. cDNA was hybridised to the Affymetrix Clariom S microarray and data analysed using R/Bioconductor. Differential expression of genes was assessed using empirical Bayes and moderated‐<jats:italic>t</jats:italic>‐statistic approaches. Category enrichment analysis was used with a tissue‐specific background to examine the biological context of differentially expressed genes. Selected differentially regulated genes were validated by qPCR. Enzyme‐linked immunosorbent assay (ELISA) for intelectin‐1 was performed on all VAT samples. The previously‐described cohort plus 12 additional patients from each group also had plasma I = intelectin‐1 ELISA carried out.</jats:p></jats:sec><jats:sec><jats:title>Results</jats:title><jats:p>In VAT vs. SAT comparisons, there were 2101, 1722, and 1659 significantly regulated genes in the cachectic, weight stable, and control groups, respectively. There were 2200 significantly regulated genes from VAT in cachectic patients compared with controls. Genes involving inflammation were enriched in cancer and control VAT vs. SAT, although different genes contributed to enrichment in each group. Energy metabolism, fat browning (e.g. uncoupling protein 1), and adipogenesis genes were down‐regulated in cancer VAT <jats:italic>(P</jats:italic> = 0.043, <jats:italic>P</jats:italic> = 5.4 × 10<jats:sup>−6</jats:sup> and <jats:italic>P</jats:italic> = 1 × 10<jats:sup>−6</jats:sup> respectively). The gene showing the largest difference in expression was ITLN1, the gene that encodes for intelectin‐1 (false discovery rate‐corrected <jats:italic>P</jats:italic> = 0.0001), a novel adipocytokine associated with weight loss in other contexts.</jats:p></jats:sec><jats:sec><jats:title>Conclusions</jats:title><jats:p>SAT and VAT have unique gene expression signatures in cancer and cachexia. VAT is metabolically active in cancer, and intelectin‐1 may be a target for therapeutic manipulation. VAT may play a fundamental role in cachexia, but the down‐regulation of energy metabolism genes implies a limited role for fat browning in cachectic patients, in contrast to pre‐clinical models.</jats:p></jats:sec>
container_issue 4
container_start_page 1141
container_title Journal of Cachexia, Sarcopenia and Muscle
container_volume 11
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792343562244849665
geogr_code not assigned
last_indexed 2024-03-01T16:53:37.417Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Adipose+depot+gene+expression+and+intelectin%E2%80%901+in+the+metabolic+response+to+cancer+and+cachexia&rft.date=2020-08-01&genre=article&issn=2190-6009&volume=11&issue=4&spage=1141&epage=1153&pages=1141-1153&jtitle=Journal+of+Cachexia%2C+Sarcopenia+and+Muscle&atitle=Adipose+depot+gene+expression+and+intelectin%E2%80%901+in+the+metabolic+response+to+cancer+and+cachexia&aulast=Skipworth&aufirst=Richard+J.E.&rft_id=info%3Adoi%2F10.1002%2Fjcsm.12568&rft.language%5B0%5D=eng
SOLR
_version_ 1792343562244849665
author Miller, Janice, Dreczkowski, Gillian, Ramage, Michael I., Wigmore, Stephen J., Gallagher, Iain J., Skipworth, Richard J.E.
author_facet Miller, Janice, Dreczkowski, Gillian, Ramage, Michael I., Wigmore, Stephen J., Gallagher, Iain J., Skipworth, Richard J.E., Miller, Janice, Dreczkowski, Gillian, Ramage, Michael I., Wigmore, Stephen J., Gallagher, Iain J., Skipworth, Richard J.E.
author_sort miller, janice
container_issue 4
container_start_page 1141
container_title Journal of Cachexia, Sarcopenia and Muscle
container_volume 11
description <jats:title>Abstract</jats:title><jats:sec><jats:title>Background</jats:title><jats:p>Cancer cachexia is a poorly understood metabolic consequence of cancer. During cachexia, different adipose depots demonstrate differential wasting rates. Animal models suggest adipose tissue may be a key driver of muscle wasting through fat–muscle crosstalk, but human studies in this area are lacking. We performed global gene expression profiling of visceral (VAT) and subcutaneous (SAT) adipose from weight stable and cachectic cancer patients and healthy controls.</jats:p></jats:sec><jats:sec><jats:title>Methods</jats:title><jats:p>Cachexia was defined as &gt;2% weight loss plus low computed tomography‐muscularity. Biopsies of SAT and VAT were taken from patients undergoing resection for oesophago‐gastric cancer, and healthy controls (<jats:italic>n</jats:italic> = 16 and 8 respectively). RNA was isolated and reverse transcribed. cDNA was hybridised to the Affymetrix Clariom S microarray and data analysed using R/Bioconductor. Differential expression of genes was assessed using empirical Bayes and moderated‐<jats:italic>t</jats:italic>‐statistic approaches. Category enrichment analysis was used with a tissue‐specific background to examine the biological context of differentially expressed genes. Selected differentially regulated genes were validated by qPCR. Enzyme‐linked immunosorbent assay (ELISA) for intelectin‐1 was performed on all VAT samples. The previously‐described cohort plus 12 additional patients from each group also had plasma I = intelectin‐1 ELISA carried out.</jats:p></jats:sec><jats:sec><jats:title>Results</jats:title><jats:p>In VAT vs. SAT comparisons, there were 2101, 1722, and 1659 significantly regulated genes in the cachectic, weight stable, and control groups, respectively. There were 2200 significantly regulated genes from VAT in cachectic patients compared with controls. Genes involving inflammation were enriched in cancer and control VAT vs. SAT, although different genes contributed to enrichment in each group. Energy metabolism, fat browning (e.g. uncoupling protein 1), and adipogenesis genes were down‐regulated in cancer VAT <jats:italic>(P</jats:italic> = 0.043, <jats:italic>P</jats:italic> = 5.4 × 10<jats:sup>−6</jats:sup> and <jats:italic>P</jats:italic> = 1 × 10<jats:sup>−6</jats:sup> respectively). The gene showing the largest difference in expression was ITLN1, the gene that encodes for intelectin‐1 (false discovery rate‐corrected <jats:italic>P</jats:italic> = 0.0001), a novel adipocytokine associated with weight loss in other contexts.</jats:p></jats:sec><jats:sec><jats:title>Conclusions</jats:title><jats:p>SAT and VAT have unique gene expression signatures in cancer and cachexia. VAT is metabolically active in cancer, and intelectin‐1 may be a target for therapeutic manipulation. VAT may play a fundamental role in cachexia, but the down‐regulation of energy metabolism genes implies a limited role for fat browning in cachectic patients, in contrast to pre‐clinical models.</jats:p></jats:sec>
doi_str_mv 10.1002/jcsm.12568
facet_avail Online, Free
finc_class_facet Biologie, Medizin
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi9qY3NtLjEyNTY4
imprint Wiley, 2020
imprint_str_mv Wiley, 2020
institution DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1
issn 2190-5991, 2190-6009
issn_str_mv 2190-5991, 2190-6009
language English
last_indexed 2024-03-01T16:53:37.417Z
match_str miller2020adiposedepotgeneexpressionandintelectin1inthemetabolicresponsetocancerandcachexia
mega_collection Wiley (CrossRef)
physical 1141-1153
publishDate 2020
publishDateSort 2020
publisher Wiley
record_format ai
recordtype ai
series Journal of Cachexia, Sarcopenia and Muscle
source_id 49
spelling Miller, Janice Dreczkowski, Gillian Ramage, Michael I. Wigmore, Stephen J. Gallagher, Iain J. Skipworth, Richard J.E. 2190-5991 2190-6009 Wiley Physiology (medical) Orthopedics and Sports Medicine http://dx.doi.org/10.1002/jcsm.12568 <jats:title>Abstract</jats:title><jats:sec><jats:title>Background</jats:title><jats:p>Cancer cachexia is a poorly understood metabolic consequence of cancer. During cachexia, different adipose depots demonstrate differential wasting rates. Animal models suggest adipose tissue may be a key driver of muscle wasting through fat–muscle crosstalk, but human studies in this area are lacking. We performed global gene expression profiling of visceral (VAT) and subcutaneous (SAT) adipose from weight stable and cachectic cancer patients and healthy controls.</jats:p></jats:sec><jats:sec><jats:title>Methods</jats:title><jats:p>Cachexia was defined as &gt;2% weight loss plus low computed tomography‐muscularity. Biopsies of SAT and VAT were taken from patients undergoing resection for oesophago‐gastric cancer, and healthy controls (<jats:italic>n</jats:italic> = 16 and 8 respectively). RNA was isolated and reverse transcribed. cDNA was hybridised to the Affymetrix Clariom S microarray and data analysed using R/Bioconductor. Differential expression of genes was assessed using empirical Bayes and moderated‐<jats:italic>t</jats:italic>‐statistic approaches. Category enrichment analysis was used with a tissue‐specific background to examine the biological context of differentially expressed genes. Selected differentially regulated genes were validated by qPCR. Enzyme‐linked immunosorbent assay (ELISA) for intelectin‐1 was performed on all VAT samples. The previously‐described cohort plus 12 additional patients from each group also had plasma I = intelectin‐1 ELISA carried out.</jats:p></jats:sec><jats:sec><jats:title>Results</jats:title><jats:p>In VAT vs. SAT comparisons, there were 2101, 1722, and 1659 significantly regulated genes in the cachectic, weight stable, and control groups, respectively. There were 2200 significantly regulated genes from VAT in cachectic patients compared with controls. Genes involving inflammation were enriched in cancer and control VAT vs. SAT, although different genes contributed to enrichment in each group. Energy metabolism, fat browning (e.g. uncoupling protein 1), and adipogenesis genes were down‐regulated in cancer VAT <jats:italic>(P</jats:italic> = 0.043, <jats:italic>P</jats:italic> = 5.4 × 10<jats:sup>−6</jats:sup> and <jats:italic>P</jats:italic> = 1 × 10<jats:sup>−6</jats:sup> respectively). The gene showing the largest difference in expression was ITLN1, the gene that encodes for intelectin‐1 (false discovery rate‐corrected <jats:italic>P</jats:italic> = 0.0001), a novel adipocytokine associated with weight loss in other contexts.</jats:p></jats:sec><jats:sec><jats:title>Conclusions</jats:title><jats:p>SAT and VAT have unique gene expression signatures in cancer and cachexia. VAT is metabolically active in cancer, and intelectin‐1 may be a target for therapeutic manipulation. VAT may play a fundamental role in cachexia, but the down‐regulation of energy metabolism genes implies a limited role for fat browning in cachectic patients, in contrast to pre‐clinical models.</jats:p></jats:sec> Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia Journal of Cachexia, Sarcopenia and Muscle
spellingShingle Miller, Janice, Dreczkowski, Gillian, Ramage, Michael I., Wigmore, Stephen J., Gallagher, Iain J., Skipworth, Richard J.E., Journal of Cachexia, Sarcopenia and Muscle, Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia, Physiology (medical), Orthopedics and Sports Medicine
title Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_full Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_fullStr Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_full_unstemmed Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_short Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_sort adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
title_unstemmed Adipose depot gene expression and intelectin‐1 in the metabolic response to cancer and cachexia
topic Physiology (medical), Orthopedics and Sports Medicine
url http://dx.doi.org/10.1002/jcsm.12568