4.7 Article

Regulation of lymphatic function and injury by nitrosative stress in obese mice

Journal

MOLECULAR METABOLISM
Volume 42, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.molmet.2020.101081

Keywords

Obesity; Lymphatic function; iNOS; Nitrosative stress; Metabolism; Inflammation

Funding

  1. NIH [R01 HL111130-01, R21 CA194882]
  2. NIH/NCI Cancer Center Support Grant [P30 CA008748]

Ask authors/readers for more resources

Objective: Obesity results in lymphatic dysfunction, but the cellular mechanisms that mediate this effect remain largely unknown. Previous studies in obese mice have shown that inducible nitric oxide synthase-expressing (iNOSthorn) inflammatory cells accumulate around lymphatic vessels. In the current study, we therefore tested the hypothesis that increased expression of iNOS results in nitrosative stress and injury to the lymphatic endothelial cells (LECs). In addition, we tested the hypothesis that lymphatic injury, independent of obesity, can modulate glucose and lipid metabolism. Methods: We compared the metabolic changes and lymphatic function of wild-type and iNOS knockout mice fed a normal chow or high-fat diet for 16 weeks. To corroborate our in vivo findings, we analyzed the effects of reactive nitrogen species on isolated LECs. Finally, using a genetically engineered mouse model that allows partial ablation of the lymphatic system, we studied the effects of acute lymphatic injury on glucose and lipid metabolism in lean mice. Results: The mesenteric lymphatic vessels of obese wild-type animals were dilated, leaky, and surrounded by iNOSthorn inflammatory cells with resulting increased accumulation of reactive nitrogen species when compared with lean wild-type or obese iNOS knockout animals. These changes in obese wild-type mice were associated with systemic glucose and lipid abnormalities, as well as decreased mesenteric LEC expression of lymphatic-specific genes, including vascular endothelial growth factor receptor 3 (VEGFR-3) and antioxidant genes as compared with lean wild type or obese iNOS knockout animals. In vitro experiments demonstrated that isolated LECs were more sensitive to reactive nitrogen species than blood endothelial cells, and that this sensitivity was ameliorated by antioxidant therapies. Finally, using mice in which the lymphatics were specifically ablated using diphtheria toxin, we found that the interaction between metabolic abnormalities caused by obesity and lymphatic dysfunction is bidirectional. Targeted partial ablation of mesenteric lymphatic channels of lean mice resulted in increased accumulation of iNOSthorn inflammatory cells and increased reactive nitrogen species. Lymphatic ablation also caused marked abnormalities in insulin sensitivity, serum glucose and insulin concentrations, expression of insulin-sensitive genes, lipid metabolism, and significantly increased systemic and mesenteric white adipose tissue (M-WAT) inflammatory responses. Conclusions: Our studies suggest that increased iNOS production in obese animals plays a key role in regulating lymphatic injury by increasing nitrosative stress. In addition, our studies suggest that obesity-induced lymphatic injury may amplify metabolic abnormalities by increasing systemic and local inflammatory responses and regulating insulin sensitivity. These findings suggest that manipulation of the lymphatic system may represent a novel means of treating metabolic abnormalities associated with obesity. (c) 2020 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Surgery

Baseline Lymphatic Dysfunction Amplifies the Negative Effects of Lymphatic Injury

Geoffrey E. Hespe, Catherine L. Ly, Raghu P. Kataru, Babak J. Mehrara

PLASTIC AND RECONSTRUCTIVE SURGERY (2019)

Article Surgery

Small Numbers of CD4+ T Cells Can Induce Development of Lymphedema

Catherine L. Ly, Daniel A. Cuzzone, Raghu P. Kataru, Babak J. Mehrara

PLASTIC AND RECONSTRUCTIVE SURGERY (2019)

Article Medical Laboratory Technology

T helper 2 differentiation is necessary for development of lymphedema

Catherine L. Ly, Gabriela D. Garcia Nores, Raghu P. Kataru, Babak J. Mehrara

TRANSLATIONAL RESEARCH (2019)

Article Multidisciplinary Sciences

Stem cell-driven lymphatic remodeling coordinates tissue regeneration

Shiri Gur-Cohen, Hanseul Yang, Sanjeethan C. Baksh, Yuxuan Miao, John Levorse, Raghu P. Kataru, Xiaolei Liu, June de la Cruz-Racelis, Babak J. Mehrara, Elaine Fuchs

SCIENCE (2019)

Article Cell Biology

Lymphatic blood filling in CLEC-2-deficient mouse models

Elizabeth J. Haining, Kate L. Lowe, Surasak Wichaiyo, Raghu P. Kataru, Zoltan Nagy, Dean Pj Kavanagh, Sian Lax, Ying Di, Bernhard Nieswandt, Benoit Ho-Tin-Noe, Babak J. Mehrara, Yotis A. Senis, Julie Rayes, Steve P. Watson

Summary: The study found that CLEC-2 expression on platelets is not necessary to maintain a barrier between the blood and lymphatic systems in unchallenged mice, but deficiency in CLEC-2 can lead to lymphatic vessel blood filling under conditions of chronic vascular remodeling. CLEC-2-driven platelet activation plays an essential role in vascular developmental programs.

PLATELETS (2021)

Review Biochemistry & Molecular Biology

Histopathologic Features of Lymphedema: A Molecular Review

Claire Y. Li, Raghu P. Kataru, Babak J. Mehrara

INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES (2020)

Review Physiology

Regulation of Lymphatic Function in Obesity

Raghu P. Kataru, Hyeong Ju Park, Jung Eun Baik, Claire Li, Jinyeon Shin, Babak J. Mehrara

FRONTIERS IN PHYSIOLOGY (2020)

Article Surgery

Comparing complications in irradiated and non-irradiated free-flaps in patients with bilateral immediate breast reconstruction and unilateral post-mastectomy radiotherapy

Catherine L. Ly, Sumun Khetpal, Danielle R. Heller, Susan A. Higgins, Brigid Killelea, Michael Alperovich, Tomer Avraham

Summary: This study compared irradiated free-flaps to non-irradiated internal controls in breast cancer patients undergoing reconstruction and PMRT. While PMRT may increase fibrosis in free-flaps, it does not significantly affect overall complications or revision surgeries. However, increased counseling may be necessary if internal mammary node irradiation is likely due to potential complications.

MICROSURGERY (2021)

Article Biochemistry & Molecular Biology

Lymphatic-specific intracellular modulation of receptor tyrosine kinase signaling improves lymphatic growth and function

Raghu P. Kataru, Jung Eun Baik, Hyeung Ju Park, Catherine L. Ly, Jinyeon Shin, Noa Schwartz, Theresa T. Lu, Sagrario Ortega, Babak J. Mehrara

Summary: The study shows that intracellular modulation of the VEGF-C signaling pathway can induce the formation of functional lymphatic networks without off-target effects and inflammation.

SCIENCE SIGNALING (2021)

Article Multidisciplinary Sciences

Nucleoside-modified VEGFC mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema

Daniel Szoke, Gabor Kovacs, Eva Kemecsei, Laszlo Balint, Kitti Szotak-Ajtay, Petra Aradi, Andrea Styevkone Dinnyes, Barbara L. Mui, Ying K. Tam, Thomas D. Madden, Katalin Kariko, Raghu P. Kataru, Michael J. Hope, Drew Weissman, Babak J. Mehrara, Norbert Pardi, Zoltan Jakus

Summary: Dysfunction of the lymphatic system leads to secondary lymphedema and decreases quality of life. Researchers demonstrate that delivery of nucleoside-modified Vascular Endothelial Growth Factor C (VEGFC) mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema.

NATURE COMMUNICATIONS (2021)

Article Biology

Pilot Study of Anti-Th2 Immunotherapy for the Treatment of Breast Cancer-Related Upper Extremity Lymphedema

Babak J. Mehrara, Hyeung Ju Park, Raghu P. Kataru, Jacqueline Bromberg, Michelle Coriddi, Jung Eun Baik, Jinyeon Shin, Claire Li, Michele R. Cavalli, Elizabeth M. Encarnacion, Meghan Lee, Kimberly J. Van Zee, Elyn Riedel, Joseph H. Dayan

Summary: The study aimed to examine the effect of inhibiting Th2 inflammation in lymphedema by using QBX258, a combination of IL4/13 neutralizing antibodies, and found that it improved quality of life and reduced pathological skin changes in women with breast cancer-related lymphedema. Treatment with QBX258 showed improvements in skin stiffness and quality of life measurements, particularly in physical and social aspects, with histological evidence of decreased epidermal changes and cytokine expression in lymphedematous skin.

BIOLOGY-BASEL (2021)

Review Immunology

Regulation of Immune Function by the Lymphatic System in Lymphedema

Raghu P. Kataru, Jung Eun Baik, Hyeung Ju Park, Itay Wiser, Sonia Rehal, Jin Yeon Shin, Babak J. Mehrara

FRONTIERS IN IMMUNOLOGY (2019)

Article Endocrinology & Metabolism

Extracellular macrophage migration inhibitory factor (MIF) downregulates adipose hormone-sensitive lipase (HSL) and contributes to obesity

Liujun Chen, Lisha Li, Donghong Cui, Yiheng Huang, Haibin Tong, Haleh Zabihi, Shuxia Wang, Yadan Qi, Ted Lakowski, Lin Leng, Suixin Liu, Hong Wu, Lawrence H. Young, Richard Bucala, Dake Qi

Summary: Attenuation of adipose hormone sensitive lipase (HSL) may impair lipolysis and exacerbate obesity. Cytokine macrophage migration inhibitory factor (MIF) plays a role in regulating adipose HSL and adipocyte hypertrophy. Both intracellular and extracellular MIF have opposing effects on HSL, but extracellular action predominates to downregulate HSL and exacerbate obesity development during high-fat diet (HFD).

MOLECULAR METABOLISM (2024)

Article Endocrinology & Metabolism

GPSM1 in POMC neurons impairs brown adipose tissue thermogenesis and provokes diet-induced obesity

Mengyang Tang, Yi Zhang, Rong Zhang, Yuemei Zhang, Jiangfei Zheng, Daixi Wang, Xinyu Wang, Jing Yan, Cheng Hu

Summary: This study aimed to explore the role of GPSM1 in POMC neurons and the underlying mechanisms in metabolic homeostasis. Through various molecular, biochemical, immunofluorescent, immunohistochemical analyses, and cell culture studies, the study revealed the pathophysiological role of GPSM1 in POMC neurons and its regulation of POMC neuron activity.

MOLECULAR METABOLISM (2024)