1
|
Jian Y, Li Y, Zhang Y, Tang M, Deng M, Liu C, Cheng M, Xiao S, Deng C, Wei Z. Lymphangiogenesis: novel strategies to promote cutaneous wound healing. BURNS & TRAUMA 2024; 12:tkae040. [PMID: 39328366 PMCID: PMC11427083 DOI: 10.1093/burnst/tkae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 09/28/2024]
Abstract
The cutaneous lymphatic system regulates tissue inflammation, fluid balance and immunological responses. Lymphangiogenesis or lymphatic dysfunction may lead to lymphedema, immune deficiency, chronic inflammation etc. Tissue regeneration and healing depend on angiogenesis and lymphangiogenesis during wound healing. Tissue oedema and chronic inflammation can slow wound healing due to impaired lymphangiogenesis or lymphatic dysfunction. For example, impaired lymphangiogenesis or lymphatic dysfunction has been detected in nonhealing wounds such as diabetic ulcers, venous ulcers and bedsores. This review summarizes the structure and function of the cutaneous lymphatic vessel system and lymphangiogenesis in wounds. Furthermore, we review wound lymphangiogenesis processes and remodelling, especially the influence of the inflammatory phase. Finally, we outline how to control lymphangiogenesis to promote wound healing, assess the possibility of targeting lymphangiogenesis as a novel treatment strategy for chronic wounds and provide an analysis of the possible problems that need to be addressed.
Collapse
Affiliation(s)
- Yang Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanqi Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanji Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingyuan Tang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingfu Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Chenxiaoxiao Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Maolin Cheng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| |
Collapse
|
2
|
Jeong HH, Kim D, Kim T, Suh HP, Pak CJ, Jeon JY, Hong JP. The Role of Escin as a Topical Agent for Lymphedema Treatment in a Rat Model. INT J LOW EXTR WOUND 2023:15347346231195944. [PMID: 37605640 DOI: 10.1177/15347346231195944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Escin, a naturally derived material isolated from horse chestnut, is used as an anti-inflammatory and anti-edema agent. This study aimed to evaluate its effects on lymphedema in a rat tail model. We divided the rats into five groups. The treatment groups received topical application of escin gel at concentrations of 20%, 10%, 2%, and 0.5% for 4 weeks. The fifth group served as a control. We performed volumetric (water displacement) tests, H&E staining, and LYVE-1 immunohistochemical staining, followed by statistical evaluation. All treatment groups showed significant volumetric reductions compared with the control group, but no significant differences were observed between the treatment groups. H&E staining showed a significant reduction in dermal thickness in the 20%, 10%, and 2% escin treatment groups compared to the control group. Within the treatment groups, the 2% escin group showed a significant difference compared with the 20% and 10% escin groups (p = 0.021 for both). LYVE-1 immunohistochemical staining revealed a significantly higher mean lymphatic vessel count in the 2% escin group compared with the 20%, 10%, and 0.5% escin-treated groups and the control group (p = 0.019, p = 0.025, p = 0.019, and p = 0.032 respectively). Topical escin applied to a rat tail model of acute lymphedema resulted in a significant reduction in tail volume, reduced dermal thickness, and increased lymphatic structures. The 2% escin concentration may be the optimal dose for improving lymphedema in this model. Further research is warranted to explore the clinical application of escin in patients with lymphedema.
Collapse
Affiliation(s)
- Hyung Hwa Jeong
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| | - Donggeun Kim
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| | - Taehyun Kim
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| | - HyunSuk Peter Suh
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| | - ChangSik John Pak
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| | - Jae Yong Jeon
- Department of Rehabilitation Medicine, Asan Medical Center. University of Ulsan Collage of Medicine, Seoul, Republic of Korea
| | - Joon Pio Hong
- Department of Plastic and Reconstructive Surgery, Asan Medical Center, University of Ulsan, School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Promotion of Lymphangiogenesis by Targeted Delivery of VEGF-C Improves Diabetic Wound Healing. Cells 2023; 12:cells12030472. [PMID: 36766814 PMCID: PMC9913977 DOI: 10.3390/cells12030472] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Chronic wounds represent a major therapeutic challenge. Lymphatic vessel function is impaired in chronic ulcers but the role of lymphangiogenesis in wound healing has remained unclear. We found that lymphatic vessels are largely absent from chronic human wounds as evaluated in patient biopsies. Excisional wound healing studies were conducted using transgenic mice with or without an increased number of cutaneous lymphatic vessels, as well as antibody-mediated inhibition of lymphangiogenesis. We found that a lack of lymphatic vessels mediated a proinflammatory wound microenvironment and delayed wound closure, and that the VEGF-C/VEGFR3 signaling axis is required for wound lymphangiogenesis. Treatment of diabetic mice (db/db mice) with the F8-VEGF-C fusion protein that targets the alternatively spliced extra domain A (EDA) of fibronectin, expressed in remodeling tissue, promoted wound healing, and potently induced wound lymphangiogenesis. The treatment also reduced tissue inflammation and exerted beneficial effects on the wound microenvironment, including myofibroblast density and collagen deposition. These findings indicate that activating the lymphatic vasculature might represent a new therapeutic strategy for treating chronic non-healing wounds.
Collapse
|
4
|
Kong AM, Lim SY, Palmer JA, Rixon A, Gerrand YW, Yap KK, Morrison WA, Mitchell GM. Engineering transplantable human lymphatic and blood capillary networks in a porous scaffold. J Tissue Eng 2022; 13:20417314221140979. [PMID: 36600999 PMCID: PMC9806376 DOI: 10.1177/20417314221140979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/08/2022] [Indexed: 12/27/2022] Open
Abstract
Due to a relative paucity of studies on human lymphatic assembly in vitro and subsequent in vivo transplantation, capillary formation and survival of primary human lymphatic (hLEC) and blood endothelial cells (hBEC) ± primary human vascular smooth muscle cells (hvSMC) were evaluated and compared in vitro and in vivo. hLEC ± hvSMC or hBEC ± hvSMC were seeded in a 3D porous scaffold in vitro, and capillary percent vascular volume (PVV) and vascular density (VD)/mm2 assessed. Scaffolds were also transplanted into a sub-cutaneous rat wound with morphology/morphometry assessment. Initially hBEC formed a larger vessel network in vitro than hLEC, with interconnected capillaries evident at 2 days. Interconnected lymphatic capillaries were slower (3 days) to assemble. hLEC capillaries demonstrated a significant overall increase in PVV (p = 0.0083) and VD (p = 0.0039) in vitro when co-cultured with hvSMC. A similar increase did not occur for hBEC + hvSMC in vitro, but hBEC + hvSMC in vivo significantly increased PVV (p = 0.0035) and VD (p = 0.0087). Morphology/morphometry established that hLEC vessels maintained distinct cell markers, and demonstrated significantly increased individual vessel and network size, and longer survival than hBEC capillaries in vivo, and established inosculation with rat lymphatics, with evidence of lymphatic function. The porous polyurethane scaffold provided advantages to capillary network formation due to its large (300-600 μm diameter) interconnected pores, and sufficient stability to ensure successful surgical transplantation in vivo. Given their successful survival and function in vivo within the porous scaffold, in vitro assembled hLEC networks using this method are potentially applicable to clinical scenarios requiring replacement of dysfunctional or absent lymphatic networks.
Collapse
Affiliation(s)
- Anne M Kong
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Shiang Y Lim
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Drug Discovery Biology, Faculty of
Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC,
Australia
- National Heart Research Institute
Singapore, National Heart Centre Singapore
| | - Jason A Palmer
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Centre for Eye Research Australia, East
Melbourne, VIC, Australia
| | - Amanda Rixon
- Experimental Medical and Surgical Unit,
St Vincent’s Hospital Melbourne, Fitzroy, VIC, Australia
| | - Yi-Wen Gerrand
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Kiryu K Yap
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
| | - Wayne A Morrison
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Faculty of Health Sciences, Australian
Catholic University, East Melbourne VIC, Australia
- Department of Plastic and
Reconstructive Surgery, St Vincent’s Hospital Melbourne, Fitzroy, VIC,
Australia
| | - Geraldine M Mitchell
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Faculty of Health Sciences, Australian
Catholic University, East Melbourne VIC, Australia
- Geraldine M Mitchell, O’Brien Institute
Department at St Vincent’s Institute of Medical Research, 9 Princes Street,
Fitzroy, VIC 3065, Australia.
| |
Collapse
|
5
|
Baik JE, Park HJ, Kataru RP, Savetsky IL, Ly CL, Shin J, Encarnacion EM, Cavali MR, Klang MG, Riedel E, Coriddi M, Dayan JH, Mehrara BJ. TGF-β1 mediates pathologic changes of secondary lymphedema by promoting fibrosis and inflammation. Clin Transl Med 2022; 12:e758. [PMID: 35652284 PMCID: PMC9160979 DOI: 10.1002/ctm2.758] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/15/2022] Open
Abstract
Background Secondary lymphedema is a common complication of cancer treatment, and previous studies have shown that the expression of transforming growth factor‐beta 1 (TGF‐β1), a pro‐fibrotic and anti‐lymphangiogenic growth factor, is increased in this disease. Inhibition of TGF‐β1 decreases the severity of the disease in mouse models; however, the mechanisms that regulate this improvement remain unknown. Methods Expression of TGF‐β1 and extracellular matrix molecules (ECM) was assessed in biopsy specimens from patients with unilateral breast cancer‐related lymphedema (BCRL). The effects of TGF‐β1 inhibition using neutralizing antibodies or a topical formulation of pirfenidone (PFD) were analyzed in mouse models of lymphedema. We also assessed the direct effects of TGF‐β1 on lymphatic endothelial cells (LECs) using transgenic mice that expressed a dominant‐negative TGF‐β receptor selectively on LECs (LECDN‐RII). Results The expression of TGF‐β1 and ECM molecules is significantly increased in BCRL skin biopsies. Inhibition of TGF‐β1 in mouse models of lymphedema using neutralizing antibodies or with topical PFD decreased ECM deposition, increased the formation of collateral lymphatics, and inhibited infiltration of T cells. In vitro studies showed that TGF‐β1 in lymphedematous tissues increases fibroblast, lymphatic endothelial cell (LEC), and lymphatic smooth muscle cell stiffness. Knockdown of TGF‐β1 responsiveness in LECDN‐RII resulted in increased lymphangiogenesis and collateral lymphatic formation; however, ECM deposition and fibrosis persisted, and the severity of lymphedema was indistinguishable from controls. Conclusions Our results show that TGF‐β1 is an essential regulator of ECM deposition in secondary lymphedema and that inhibition of this response is a promising means of treating lymphedema.
Collapse
Affiliation(s)
- Jung Eun Baik
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hyeung Ju Park
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raghu P Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ira L Savetsky
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine L Ly
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jinyeon Shin
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elizabeth M Encarnacion
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michele R Cavali
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark G Klang
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elyn Riedel
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michelle Coriddi
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph H Dayan
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Babak J Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
6
|
Ishii M, Miyata H, Ikeda N, Tagawa T, Nishimura M. Piper retrofractum extract and its component piperine promote lymphangiogenesis via an AKT- and ERK-dependent mechanism. J Food Biochem 2022; 46:e14233. [PMID: 35567300 DOI: 10.1111/jfbc.14233] [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: 03/08/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022]
Abstract
Administration of Piper retrofractum extract (PRE) has been reported to alleviate edema, but the mechanism underlying this effect is unknown. Promotion of lymphangiogenesis is known to improve lymphedema, but the effect of PRE on lymphangiogenesis remains unclear. In the present study, we investigated whether PRE and specifically, piperine, the main component of PRE, can induce lymphangiogenesis. Treatments with PRE and piperine significantly promoted the proliferation, migration, and tube formation in human dermal lymphatic microvascular endothelial cells (HDLECs) but had no effect on the expression of lymphangiogenic factors. Furthermore, PRE and piperine significantly promoted the phosphorylation of the AKT and ERK proteins in HDLECs, and pretreatment with AKT and ERK inhibitors significantly attenuated the PRE- and piperine-induced lymphangiogenesis. These results indicate that PRE and piperine promote lymphangiogenesis via an AKT- and ERK-dependent mechanism. PRACTICAL APPLICATIONS: The lymphatic system plays various roles such as maintaining tissue fluid homeostasis, immune defense, and metabolism. Disruption of the lymphatic system results in insufficient fluid drainage, which causes edema. Currently, there are no effective treatments for lymphedema; therefore, the development of novel treatment strategies is desirable. In this study, we showed that PRE and its main component piperine promote lymphangiogenesis in lymphatic endothelial cells. Therefore, PRE has the potential to be used as a novel functional food for relieving lymphedema.
Collapse
Affiliation(s)
- Masakazu Ishii
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Haruka Miyata
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Nao Ikeda
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Masahiro Nishimura
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
7
|
Szőke D, Kovács G, Kemecsei É, Bálint L, Szoták-Ajtay K, Aradi P, Styevkóné Dinnyés A, Mui BL, Tam YK, Madden TD, Karikó K, Kataru RP, Hope MJ, Weissman D, Mehrara BJ, Pardi N, Jakus Z. Nucleoside-modified VEGFC mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema. Nat Commun 2021; 12:3460. [PMID: 34103491 PMCID: PMC8187400 DOI: 10.1038/s41467-021-23546-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/30/2021] [Indexed: 12/01/2022] Open
Abstract
Lack or dysfunction of the lymphatics leads to secondary lymphedema formation that seriously reduces the function of the affected organs and results in degradation of quality of life. Currently, there is no definitive treatment option for lymphedema. Here, we utilized nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNPs) encoding murine Vascular Endothelial Growth Factor C (VEGFC) to stimulate lymphatic growth and function and reduce experimental lymphedema in mouse models. We demonstrated that administration of a single low-dose of VEGFC mRNA-LNPs induced durable, organ-specific lymphatic growth and formation of a functional lymphatic network. Importantly, VEGFC mRNA-LNP treatment reversed experimental lymphedema by restoring lymphatic function without inducing any obvious adverse events. Collectively, we present a novel application of the nucleoside-modified mRNA-LNP platform, describe a model for identifying the organ-specific physiological and pathophysiological roles of the lymphatics, and propose an efficient and safe treatment option that may serve as a novel therapeutic tool to reduce lymphedema.
Collapse
Affiliation(s)
- Dániel Szőke
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Gábor Kovács
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Éva Kemecsei
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - László Bálint
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Kitti Szoták-Ajtay
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Petra Aradi
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Andrea Styevkóné Dinnyés
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | | | | | - Raghu P Kataru
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Drew Weissman
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Babak J Mehrara
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Norbert Pardi
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.
- MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary.
| |
Collapse
|
8
|
Abstract
Tissue engineering has witnessed remarkable advancement in various fields of medicine and has the potential of revolutionizing the management of lymphedema. Combining approaches of biotechnology with the evolving understanding of lymphangiogenesis may offer promising treatment modalities for patients suffering from lymphedema. The strategies to lymphatic vessels tissue engineer can be grouped into four main categories: Delivery of chemokines, cytokines, and other growth factors to induce lymphangiogenesis; cell-based approach using lymphatic endothelial cells or stem-cells; scaffold-based tissue engineering; or a combination of these. This review will summarize the current approach to cancer-related lymphedema and advances in lymphatic tissue engineering strategies and the challenges facing the regeneration of lymphatic vasculature, particularly in an oncologic setting.
Collapse
Affiliation(s)
- Malke Asaad
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Summer E Hanson
- Section of Plastic and Reconstructive Surgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| |
Collapse
|
9
|
Jia W, Hitchcock-Szilagyi H, He W, Goldman J, Zhao F. Engineering the Lymphatic Network: A Solution to Lymphedema. Adv Healthc Mater 2021; 10:e2001537. [PMID: 33502814 PMCID: PMC8483563 DOI: 10.1002/adhm.202001537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/06/2020] [Indexed: 12/18/2022]
Abstract
Secondary lymphedema is a life-long disorder characterized by chronic tissue swelling and inflammation that obstruct interstitial fluid circulation and immune cell trafficking. Regenerating lymphatic vasculatures using various strategies represents a promising treatment for lymphedema. Growth factor injection and gene delivery have been developed to stimulate lymphangiogenesis and augment interstitial fluid resorption. Using bioengineered materials as growth factor delivery vehicles allows for a more precisely targeted lymphangiogenic activation within the injured site. The implantation of prevascularized lymphatic tissue also promotes in situ lymphatic capillary network formation. The engineering of larger scale lymphatic tissues, including lymphatic collecting vessels and lymph nodes constructed by bioengineered scaffolds or decellularized animal tissues, offers alternatives to reconnecting damaged lymphatic vessels and restoring lymph circulation. These approaches provide lymphatic vascular grafting materials to reimpose lymphatic continuity across the site of injury, without creating secondary injuries at donor sites. The present work reviews molecular mechanisms mediating lymphatic system development, approaches to promoting lymphatic network regeneration, and strategies for engineering lymphatic tissues, including lymphatic capillaries, collecting vessels, and nodes. Challenges of advanced translational applications are also discussed.
Collapse
Affiliation(s)
- Wenkai Jia
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77845
| | | | - Weilue He
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77845
| |
Collapse
|
10
|
Künnapuu J, Bokharaie H, Jeltsch M. Proteolytic Cleavages in the VEGF Family: Generating Diversity among Angiogenic VEGFs, Essential for the Activation of Lymphangiogenic VEGFs. BIOLOGY 2021; 10:167. [PMID: 33672235 PMCID: PMC7926383 DOI: 10.3390/biology10020167] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/24/2022]
Abstract
Specific proteolytic cleavages turn on, modify, or turn off the activity of vascular endothelial growth factors (VEGFs). Proteolysis is most prominent among the lymph-angiogenic VEGF-C and VEGF-D, which are synthesized as precursors that need to undergo enzymatic removal of their C- and N-terminal propeptides before they can activate their receptors. At least five different proteases mediate the activating cleavage of VEGF-C: plasmin, ADAMTS3, prostate-specific antigen, cathepsin D, and thrombin. All of these proteases except for ADAMTS3 can also activate VEGF-D. Processing by different proteases results in distinct forms of the "mature" growth factors, which differ in affinity and receptor activation potential. The "default" VEGF-C-activating enzyme ADAMTS3 does not activate VEGF-D, and therefore, VEGF-C and VEGF-D do function in different contexts. VEGF-C itself is also regulated in different contexts by distinct proteases. During embryonic development, ADAMTS3 activates VEGF-C. The other activating proteases are likely important for non-developmental lymphangiogenesis during, e.g., tissue regeneration, inflammation, immune response, and pathological tumor-associated lymphangiogenesis. The better we understand these events at the molecular level, the greater our chances of developing successful therapies targeting VEGF-C and VEGF-D for diseases involving the lymphatics such as lymphedema or cancer.
Collapse
Affiliation(s)
- Jaana Künnapuu
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Honey Bokharaie
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Michael Jeltsch
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Wihuri Research Institute, 00290 Helsinki, Finland
| |
Collapse
|
11
|
Abu-Rustum NR, Angioli R, Bailey AE, Broach V, Buda A, Coriddi MR, Dayan JH, Frumovitz M, Kim YM, Kimmig R, Leitao MM, Muallem MZ, McKittrick M, Mehrara B, Montera R, Moukarzel LA, Naik R, Pedra Nobre S, Plante M, Plotti F, Zivanovic O. IGCS Intraoperative Technology Taskforce. Update on near infrared imaging technology: beyond white light and the naked eye, indocyanine green and near infrared technology in the treatment of gynecologic cancers. Int J Gynecol Cancer 2020; 30:670-683. [PMID: 32234846 PMCID: PMC8867216 DOI: 10.1136/ijgc-2019-001127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Nadeem R Abu-Rustum
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | - Arthur E Bailey
- Research and Development, Stryker Endoscopy, San Jose, California, USA
| | - Vance Broach
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Alessandro Buda
- Department of Obstetrics and Gynecology, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Michelle R Coriddi
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Joseph H Dayan
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Michael Frumovitz
- Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yong Man Kim
- Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Rainer Kimmig
- Gynecology and Obstetrics, University Hospital of Duisburg-Essen, Essen, Germany
| | - Mario M Leitao
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Mustafa Zelal Muallem
- Department of Gynecology with Center for Oncological Surgery, Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Virchow Campus Clinic, Charité Medical University, Berlin, Germany
| | - Matt McKittrick
- Research and Development, Stryker Endoscopy, San Jose, California, USA
| | - Babak Mehrara
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Roberto Montera
- Universita Campus Bio-Medico di Roma Facolta di Medicina e Chirurgia, Roma, Lazio, Italy
| | - Lea A Moukarzel
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Raj Naik
- Northern Gynaecological Oncology Centre, Queen Elizabeth Hospital, Gateshead, UK
| | - Silvana Pedra Nobre
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Marie Plante
- Obstetrics and Gynecology, Centre Hospitalier Universitaire de Quebec, Quebec, Quebec, Canada
| | - Francesco Plotti
- Universita Campus Bio-Medico di Roma Facolta di Medicina e Chirurgia, Roma, Lazio, Italy
| | - Oliver Zivanovic
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
12
|
Hemostasis stimulates lymphangiogenesis through release and activation of VEGFC. Blood 2020; 134:1764-1775. [PMID: 31562136 DOI: 10.1182/blood.2019001736] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/21/2019] [Indexed: 02/07/2023] Open
Abstract
Hemostasis associated with tissue injury is followed by wound healing, a complex process by which damaged cellular material is removed and tissue repaired. Angiogenic responses are a central aspect of wound healing, including the growth of new lymphatic vessels by which immune cells, protein, and fluid are transported out of the wound area. The concept that hemostatic responses might be linked to wound healing responses is an old one, but demonstrating such a link in vivo and defining specific molecular mechanisms by which the 2 processes are connected has been difficult. In the present study, we demonstrate that the lymphangiogenic factors vascular endothelial growth factor C (VEGFC) and VEGFD are cleaved by thrombin and plasmin, serine proteases generated during hemostasis and wound healing. Using a new tail-wounding assay to test the relationship between clot formation and lymphangiogenesis in mice, we find that platelets accelerate lymphatic growth after injury in vivo. Genetic studies reveal that platelet enhancement of lymphatic growth after wounding is dependent on the release of VEGFC, but not VEGFD, a finding consistent with high expression of VEGFC in both platelets and avian thrombocytes. Analysis of lymphangiogenesis after full-thickness skin excision, a wound model that is not associated with significant clot formation, also revealed an essential role for VEGFC, but not VEGFD. These studies define a concrete molecular and cellular link between hemostasis and lymphangiogenesis during wound healing and reveal that VEGFC, the dominant lymphangiogenic factor during embryonic development, continues to play a dominant role in lymphatic growth in mature animals.
Collapse
|
13
|
Forte AJ, Boczar D, Huayllani MT, McLaughlin SA, Bagaria S. Use of Gene Transfer Vectors in Lymphedema Treatment: A Systematic Review. Cureus 2019; 11:e5887. [PMID: 31772857 PMCID: PMC6837272 DOI: 10.7759/cureus.5887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Different delivery mechanisms have been proposed in the literature for targeted therapies in the treatment of lymphedema. They vary from simple and direct injection to sophisticated induction of gene expression in a targeted tissue. We conducted a systematic review of publications assessing the use of viral vectors for gene transfer in lymphedema treatment. We hypothesized that viral vectors are an effective way to deliver targeted therapy in lymphedema treatment. We conducted a comprehensive systematic review of the published literature on targeted therapies associated with lymphedema surgery using the PubMed database. Eligibility criteria excluded papers that reported use of viral vectors for other medical conditions. Abstracts, presentations, reviews, meta-analyses, and non-English language articles were also excluded. From 21 potential articles found in the literature, fourteen fulfilled study eligibility criteria. Positive outcomes in terms of lymphangiogenesis were seen. The viral vectors used included adenovirus and recombinant adeno-associated virus. Most of the genes expressed were growth factors, but expression of dominant-negative transforming growth factor-β1 receptor-II or Prox1 was also proposed. Five studies targeted genetic expression on lymphedema tissue, five on transplanted lymph nodes, two on skeletal muscle, and one on adipose-derived stem cells. Publications assessing use of viral vectors for gene transfer in lymphedema treatment demonstrated that it is an effective mechanism of delivering targeted therapies. However, to date, all studies were experimental and further studies must be performed before translating these therapies into clinical practice.
Collapse
Affiliation(s)
- Antonio J Forte
- Plastic Surgery, Mayo Clinic Florida - Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| | - Daniel Boczar
- Plastic Surgery, Mayo Clinic Florida - Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| | - Maria T Huayllani
- Plastic Surgery, Mayo Clinic Florida - Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| | - Sarah A McLaughlin
- Surgery, Mayo Clinic Florida - Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| | - Sanjay Bagaria
- Surgery, Mayo Clinic Florida - Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Jacksonville, USA
| |
Collapse
|
14
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
15
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
16
|
Ferrão JSP, Bonfim Neto AP, da Fonseca VU, Sousa LMMDC, Papa PDC. Vascular endothelial growth factor C treatment for mouse hind limb lymphatic revascularization. Vet Med Sci 2019; 5:249-259. [PMID: 30746892 PMCID: PMC6498523 DOI: 10.1002/vms3.151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Spontaneous lymphatic revascularization is a challenge and the establishment of new therapeutic strategies may improve life quality for patients suffering from lymphatic disorders. This study was designed to verify if VEGFC treatment improves lymphatic vascularization in a time‐dependent manner in mouse hindlimb (HL) after resection of the inguinal lymph node. Lymphatic vascular density (Vv) and length (Lv) were evaluated by stereology after immunohistochemistry. The control Group (CG) was not manipulated but received saline instead of VEGFC treatment. The surgery Group (SG) had the left inguinal lymph node resected but did not received VEGFC treatment. VEGFC Treated Group (TG) had the node resected and received VEGFC treatment. VEGFC and VEGFR3 local expression were assessed by qPCR. There was an effect of time over Vv and Lv in the SG and significant difference between CG and SG in the regions studied (proximal, medium and distal regions) of the left HL (LHL). The Lv showed significant difference between CG and SG only in the medium region. The Vv and the Lv for TG were higher than the other groups. VEGFC and VEGFR3 gene expression presented time effect in all regions of the LHL for SG and TG. Both VEGFC and VEGFR3 gene expression presented significant difference between CG and SG, between SG and TG and between CG and TG. This study showed significant decrease in lymphatic vascularization in the left hindlimb of mice after surgical removal of the inguinal lymph node and adjacent lymphatic vessels. Exogenous VEGFC could recover lymphatic vascularization through stimulating neolymphangiogenesis.
Collapse
Affiliation(s)
- Juliana S P Ferrão
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Antenor P Bonfim Neto
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Vanessa U da Fonseca
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Paula de C Papa
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Kilarski WW. Physiological Perspective on Therapies of Lymphatic Vessels. Adv Wound Care (New Rochelle) 2018; 7:189-208. [PMID: 29984111 PMCID: PMC6032671 DOI: 10.1089/wound.2017.0768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/26/2018] [Indexed: 12/16/2022] Open
Abstract
Significance: Growth of distinctive blood vessels of granulation tissue is a central step in the post-developmental tissue remodeling. Even though lymphangiogenesis is a part of the regeneration process, the significance of the controlled restoration of lymphatic vessels has only recently been recognized. Recent Advances: Identification of lymphatic markers and growth factors paved the way for the exploration of the roles of lymphatic vessels in health and disease. Emerging pro-lymphangiogenic therapies use vascular endothelial growth factor (VEGF)-C to combat fluid retention disorders such as lymphedema and to enhance the local healing process. Critical Issues: The relevance of recently identified lymphatic functions awaits verification by their association with pathologic conditions. Further, despite a century of research, the complete etiology of secondary lymphedema, a fluid retention disorder directly linked to the lymphatic function, is not understood. Finally, the specificity of pro-lymphangiogenic therapy depends on VEGF-C transfection efficiency, dose exposure, and the age of the subject, factors that are difficult to standardize in a heterogeneous human population. Future Directions: Further research should reveal the role of lymphatic circulation in internal organs and connect its impairment with human diseases. Pro-lymphangiogenic therapies that aim at the acceleration of tissue healing should focus on the controlled administration of VEGF-C to increase their capillary specificity, whereas regeneration of collecting vessels might benefit from balanced maturation and differentiation of pre-existing lymphatics. Unique features of pre-nodal lymphatics, fault tolerance and functional hyperplasia of capillaries, may find applications outreaching traditional pro-lymphangiogenic therapies, such as immunomodulation or enhancement of subcutaneous grafting.
Collapse
Affiliation(s)
- Witold W. Kilarski
- Institute for Molecular Engineering, The University of Chicago, Chicago, Illinois
| |
Collapse
|
18
|
Multipotent Adult Progenitor Cells Support Lymphatic Regeneration at Multiple Anatomical Levels during Wound Healing and Lymphedema. Sci Rep 2018; 8:3852. [PMID: 29497054 PMCID: PMC5832783 DOI: 10.1038/s41598-018-21610-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/02/2018] [Indexed: 12/20/2022] Open
Abstract
Lymphatic capillary growth is an integral part of wound healing, yet, the combined effectiveness of stem/progenitor cells on lymphatic and blood vascular regeneration in wounds needs further exploration. Stem/progenitor cell transplantation also emerged as an approach to cure lymphedema, a condition caused by lymphatic system deficiency. While lymphedema treatment requires lymphatic system restoration from the capillary to the collector level, it remains undetermined whether stem/progenitor cells support a complex regenerative response across the entire anatomical spectrum of the system. Here, we demonstrate that, although multipotent adult progenitor cells (MAPCs) showed potential to differentiate down the lymphatic endothelial lineage, they mainly trophically supported lymphatic endothelial cell behaviour in vitro. In vivo, MAPC transplantation supported blood vessel and lymphatic capillary growth in wounds and restored lymph drainage across skin flaps by stimulating capillary and pre-collector vessel regeneration. Finally, human MAPCs mediated survival and functional reconnection of transplanted lymph nodes to the host lymphatic network by improving their (lymph)vascular supply and restoring collector vessels. Thus, MAPC transplantation represents a promising remedy for lymphatic system restoration at different anatomical levels and hence an appealing treatment for lymphedema. Furthermore, its combined efficacy on lymphatic and blood vascular growth is an important asset for wound healing.
Collapse
|
19
|
McLaughlin SA, Staley AC, Vicini F, Thiruchelvam P, Hutchison NA, Mendez J, MacNeill F, Rockson SG, DeSnyder SM, Klimberg S, Alatriste M, Boccardo F, Smith ML, Feldman SM. Considerations for Clinicians in the Diagnosis, Prevention, and Treatment of Breast Cancer-Related Lymphedema: Recommendations from a Multidisciplinary Expert ASBrS Panel : Part 1: Definitions, Assessments, Education, and Future Directions. Ann Surg Oncol 2017; 24:2818-2826. [PMID: 28766232 DOI: 10.1245/s10434-017-5982-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Indexed: 12/21/2022]
Affiliation(s)
| | | | - Frank Vicini
- Radiation Oncology, UCLA School of Medicine, Los Angeles, USA
| | | | - Nancy A Hutchison
- Courage Kenny Rehabilitation Institute of AllinaHealth, Minneapolis, MN, USA
| | | | - Fiona MacNeill
- Association of Breast Surgery Great Britain and Ireland, Royal College of Surgeons of England, London, UK
| | - Stanley G Rockson
- Center for Lymphatic and Venous Disorders, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah M DeSnyder
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Francesco Boccardo
- Department of Surgery, Unit of Lymphatic Surgery - S. Martino University Hospital, University of Genoa, Genoa, Italy
| | - Mark L Smith
- Hofstra Northwell School of Medicine, Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Sheldon M Feldman
- Division of Breast Surgery and Breast Surgical Oncology, Department of Surgery, Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
20
|
Güç E, Briquez PS, Foretay D, Fankhauser MA, Hubbell JA, Kilarski WW, Swartz MA. Local induction of lymphangiogenesis with engineered fibrin-binding VEGF-C promotes wound healing by increasing immune cell trafficking and matrix remodeling. Biomaterials 2017; 131:160-175. [DOI: 10.1016/j.biomaterials.2017.03.033] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 01/13/2023]
|
21
|
Li K, Min P, Sadigh P, Grassetti L, Lazzeri D, Torresetti M, Marsili R, Feng S, Liu N, Zhang YX. Prefabricated Cervical Skin Flaps for Hemi-Facial Resurfacing: Elucidating the Natural History of Postoperative Edema Using Indocyanine Green. Lymphat Res Biol 2017; 16:100-108. [PMID: 28135123 DOI: 10.1089/lrb.2015.0043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The increases in capillary wall permeability and capillary hydrostatic pressure are considered to be the causes for the acute swelling seen in flaps; however, disruption of the circulating flap lymphatics could be another contributory factor. In this study we monitor the development of flap edema in a series of 18 prefabricated flaps and aim to delineate the natural history of this phenomenon by use of lymphography. METHODS Postoperative swelling was monitored in a series of 18 pre-expanded prefabricated cervical skin flaps used for hemi-facial burns-scar resurfacing. Time to spontaneous resolution, presence or absence of venous congestion, and clinical outcome were recorded. In two cases, indocyanine-green (ICG) lymphography was used to monitor the dermal backflow pattern until swelling had completely resolved. Average moving velocity of ICG after injection as well as flap thickness was also recorded over the follow-up period. RESULTS The average moving velocity of ICG in the flap lymphatics improved from 0.48 cm/min to 1.5 cm/min in the first 12 days after flap transfer. The dermal backflow pattern was stardust in the first 12 days, indicating moderate lymphedema, transforming to splash from week three, and a robust collecting lymphatic vessel occurring from the fifth month, indicating mild lymphedema and lymphatic channel recovery, respectively. CONCLUSION Transient swelling was observed in all prefabricated flaps in our series. We postulate that this is mostly secondary to lymphatic disruption that subsides as lymphangiogenesis takes place. ICG lymphography is an inexpensive, safe, and easy-to-use imaging technology that could be used in the monitoring of postoperative lymphedema seen in prefabricated flaps.
Collapse
Affiliation(s)
- Ke Li
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, China
| | - Peiru Min
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, China
| | - Parviz Sadigh
- 2 The Royal London Hospital , Department of Plastic Reconstructive Surgery, London, United Kingdom
| | - Luca Grassetti
- 3 Department of Plastic and Reconstructive Surgery, Marche Polytechnic University Medical School, University Hospital of Ancona , Ancona, Italy
| | - Davide Lazzeri
- 4 Plastic Reconstructive and Aesthetic Surgery Unit, Villa Salaria Clinic , Rome, Italy
| | - Matteo Torresetti
- 4 Plastic Reconstructive and Aesthetic Surgery Unit, Villa Salaria Clinic , Rome, Italy
| | - Riccardo Marsili
- 5 Plastic and Reconstructive Surgery Unit, Hospital of Pisa , Pisa, Italy
| | - Shaoqing Feng
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, China
| | - Ningfei Liu
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, China
| | - Yi Xin Zhang
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, China
| |
Collapse
|
22
|
Gentileschi S, Servillo M, Garganese G, Fragomeni S, De Bonis F, Cina A, Scambia G, Salgarello M. The lymphatic superficial circumflex iliac vessels deep branch perforator flap: A new preventive approach to lower limb lymphedema after groin dissection-preliminary evidence. Microsurgery 2016; 37:564-573. [DOI: 10.1002/micr.30142] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 11/10/2016] [Accepted: 12/02/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Stefano Gentileschi
- Department of Plastic and Reconstructive Surgery; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Maria Servillo
- Department of Plastic and Reconstructive Surgery; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Giorgia Garganese
- Department of Gynecology; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Simona Fragomeni
- Department of Gynecology; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Francesca De Bonis
- Department of Plastic and Reconstructive Surgery; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Alessandro Cina
- Department of Radiology; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Giovanni Scambia
- Department of Gynecology; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| | - Marzia Salgarello
- Department of Plastic and Reconstructive Surgery; Catholic University Sacred Heart - Policlinico Agostino Gemelli; Largo Agostino Gemelli 1, Rome 00168 Italy
| |
Collapse
|
23
|
Milasan A, Dallaire F, Mayer G, Martel C. Effects of LDL Receptor Modulation on Lymphatic Function. Sci Rep 2016; 6:27862. [PMID: 27279328 PMCID: PMC4899717 DOI: 10.1038/srep27862] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is driven by the accumulation of immune cells and cholesterol in the arterial wall. Although recent studies have shown that lymphatic vessels play an important role in macrophage reverse cholesterol transport, the specific underlying mechanisms of this physiological feature remain unknown. In the current report, we sought to better characterize the lymphatic dysfunction that is associated with atherosclerosis by studying the physiological and temporal origins of this impairment. First, we assessed that athero-protected Pcsk9−/− mice exhibited improved collecting lymphatic vessel function throughout age when compared to WT mice for up to six months, while displaying enhanced expression of LDLR on lymphatic endothelial cells. Lymphatic dysfunction was present before the atherosclerotic lesion formation in a mouse model that is predisposed to develop atherosclerosis (Ldlr−/−; hApoB100+/+). This dysfunction was presumably associated with a defect in the collecting lymphatic vessels in a non-specific cholesterol- but LDLR-dependent manner. Treatment with a selective VEGFR-3 agonist rescued this impairment observed early in the onset of this arterial disease. We suggest that LDLR modulation is associated with early atherosclerosis-related lymphatic dysfunction, and bring forth a pleiotropic role for PCSK9 in lymphatic function. Our study unveils new potential therapeutic targets for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Montreal, Quebec, Canada
| | | | - Gaétan Mayer
- Laboratory of Molecular Cell Biology, Montreal Heart Institute Research Center, Quebec, Canada.,Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Montreal, Quebec, Canada
| |
Collapse
|
24
|
Jang DH, Song DH, Chang EJ, Jeon JY. Anti-inflammatory and lymphangiogenetic effects of low-level laser therapy on lymphedema in an experimental mouse tail model. Lasers Med Sci 2015; 31:289-96. [DOI: 10.1007/s10103-015-1854-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 12/11/2015] [Indexed: 01/28/2023]
|
25
|
Cui Y, Liu K, Monzon-Medina ME, Padera RF, Wang H, George G, Toprak D, Abdelnour E, D'Agostino E, Goldberg HJ, Perrella MA, Forteza RM, Rosas IO, Visner G, El-Chemaly S. Therapeutic lymphangiogenesis ameliorates established acute lung allograft rejection. J Clin Invest 2015; 125:4255-68. [PMID: 26485284 DOI: 10.1172/jci79693] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 08/28/2015] [Indexed: 01/13/2023] Open
Abstract
Lung transplantation is the only viable option for patients suffering from otherwise incurable end-stage pulmonary diseases such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Despite aggressive immunosuppression, acute rejection of the lung allograft occurs in over half of transplant recipients, and the factors that promote lung acceptance are poorly understood. The contribution of lymphatic vessels to transplant pathophysiology remains controversial, and data that directly address the exact roles of lymphatic vessels in lung allograft function and survival are limited. Here, we have shown that there is a marked decline in the density of lymphatic vessels, accompanied by accumulation of low-MW hyaluronan (HA) in mouse orthotopic allografts undergoing rejection. We found that stimulation of lymphangiogenesis with VEGF-C156S, a mutant form of VEGF-C with selective VEGFR-3 binding, alleviates an established rejection response and improves clearance of HA from the lung allograft. Longitudinal analysis of transbronchial biopsies from human lung transplant recipients demonstrated an association between resolution of acute lung rejection and decreased HA in the graft tissue. Taken together, these results indicate that lymphatic vessel formation after lung transplantation mediates HA drainage and suggest that treatments to stimulate lymphangiogenesis have promise for improving graft outcomes.
Collapse
|
26
|
A Study of the Post-Op Lymphedema of Prefabricated Flaps and Its Prognosis. Plast Reconstr Surg 2015. [DOI: 10.1097/01.prs.0000472415.89303.bd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Ackermann M, Wettstein R, Senaldi C, Kalbermatten DF, Konerding MA, Raffoul W, Erba P. Impact of platelet rich plasma and adipose stem cells on lymphangiogenesis in a murine tail lymphedema model. Microvasc Res 2015; 102:78-85. [PMID: 26365474 DOI: 10.1016/j.mvr.2015.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 09/06/2015] [Accepted: 09/06/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Lymphedema is an underdiagnosed pathology which in industrialized countries mainly affects cancer patients that underwent lymph node dissection and/or radiation. Currently no effective therapy is available so that patients' life quality is compromised by swellings of the concerned body region. This unfortunate condition is associated with body imbalance and subsequent osteochondral deformations and impaired function as well as with an increased risk of potentially life threatening soft tissue infections. METHODS The effects of PRP and ASC on angiogenesis (anti-CD31 staining), microcirculation (Laser Doppler Imaging), lymphangiogenesis (anti-LYVE1 staining), microvascular architecture (corrosion casting) and wound healing (digital planimetry) are studied in a murine tail lymphedema model. RESULTS Wounds treated by PRP and ASC healed faster and showed a significantly increased epithelialization mainly from the proximal wound margin. The application of PRP induced a significantly increased lymphangiogenesis while the application of ASC did not induce any significant change in this regard. CONCLUSIONS PRP and ASC affect lymphangiogenesis and lymphedema development and might represent a promising approach to improve regeneration of lymphatic vessels, restore disrupted lymphatic circulation and treat or prevent lymphedema alone or in combination with currently available lymphedema therapies.
Collapse
Affiliation(s)
- Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Reto Wettstein
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Basel, Basel, Switzerland
| | - Christopher Senaldi
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Daniel F Kalbermatten
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Basel, Basel, Switzerland
| | - Moritz A Konerding
- Institute of Functional and Clinical Anatomy, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Wassim Raffoul
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Paolo Erba
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Department of ORL, Head and Neck Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
28
|
Growth factor therapy and lymph node graft for lymphedema. J Surg Res 2015; 196:200-7. [DOI: 10.1016/j.jss.2015.02.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 01/22/2015] [Accepted: 02/13/2015] [Indexed: 12/12/2022]
|
29
|
Cell transplantation therapy for a rat model of secondary lymphedema. J Surg Res 2014; 189:184-91. [DOI: 10.1016/j.jss.2013.11.1116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/24/2013] [Accepted: 11/27/2013] [Indexed: 12/21/2022]
|
30
|
Weitman E, Cuzzone D, Mehrara BJ. Tissue engineering and regeneration of lymphatic structures. Future Oncol 2014; 9:1365-74. [PMID: 23980683 DOI: 10.2217/fon.13.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue engineering is the process by which biological structures are recreated using a combination of molecular signals, cellular components and scaffolds. Although the perceived potential of this approach to reconstruct damaged or missing tissues is seemingly limitless, application of these ideas in vivo has been more difficult than expected. However, despite these obstacles, important advancements have been reported for a number of organ systems, including recent reports on the lymphatic system. These advancements are important since the lymphatic system plays a central role in immune responses, regulation of inflammation, lipid absorption and interstitial fluid homeostasis. Insights obtained over the past two decades have advanced our understanding of the molecular and cellular mechanisms that govern lymphatic development and function. Utilizing this knowledge has led to important advancements in lymphatic tissue engineering, which is the topic of this review.
Collapse
Affiliation(s)
- Evan Weitman
- The Department of Surgery, Plastic Surgery Section, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
31
|
Choi I, Lee S, Hong YK. The new era of the lymphatic system: no longer secondary to the blood vascular system. Cold Spring Harb Perspect Med 2013; 2:a006445. [PMID: 22474611 DOI: 10.1101/cshperspect.a006445] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The blood and lymphatic systems are the two major circulatory systems in our body. Although the blood system has been studied extensively, the lymphatic system has received much less scientific and medical attention because of its elusive morphology and mysterious pathophysiology. However, a series of landmark discoveries made in the past decade has begun to change the previous misconception of the lymphatic system to be secondary to the more essential blood vascular system. In this article, we review the current understanding of the development and pathology of the lymphatic system. We hope to convince readers that the lymphatic system is no less essential than the blood circulatory system for human health and well-being.
Collapse
Affiliation(s)
- Inho Choi
- Department of Surgery, Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | |
Collapse
|
32
|
Park C, Lee JY, Yoon YS. Role of bone marrow-derived lymphatic endothelial progenitor cells for lymphatic neovascularization. Trends Cardiovasc Med 2012; 21:135-40. [PMID: 22732548 DOI: 10.1016/j.tcm.2012.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The lymphatic vasculature plays a pivotal role in maintaining tissue fluid homeostasis, immune surveillance, and lipid uptake in the gastrointestinal organs. Therefore, impaired function of the lymphatic vessels caused by genetic defects, infection, trauma, or surgery leads to the abnormal accrual of lymph fluid in the tissue and culminates in the swelling of affected tissues, known as lymphedema. Lymphedema causes impaired wound healing, compromised immune defense, and, in rare case, lymphangiosarcoma. Although millions of people suffer from lymphedema worldwide, no effective therapy is currently available. In addition, recent advances in cancer biology have disclosed an indispensable function of the lymphatic vessel in tumor growth and metastasis. Therefore, understanding the detailed mechanisms governing lymphatic vessel formation and function in pathophysiologic conditions is essential to prevent or treat these diseases. We review the developmental processes of the lymphatic vessels and postnatal lymphatic neovascularization, focusing on the role of recently identified bone marrow-derived podoplanin-expressing (podoplanin(+)) cells as lymphatic endothelial progenitor cells.
Collapse
Affiliation(s)
- Changwon Park
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | |
Collapse
|
33
|
In vivo imaging of lymphatic vessels in development, wound healing, inflammation, and tumor metastasis. Proc Natl Acad Sci U S A 2012; 109:6223-8. [PMID: 22474390 DOI: 10.1073/pnas.1115542109] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Lymphatic vessel growth or lymphangiogenesis occurs during embryonic development and wound healing and plays an important role in tumor metastasis and inflammatory diseases. However, the possibility of noninvasive detection and quantification of lymphangiogenesis has been lacking. Here, we present the Vegfr3(EGFPLuc) mouse model, where an EGFP-luciferase fusion protein, expressed under the endogenous transcriptional control of the Vegfr3 gene, allows the monitoring of physiological and pathological lymphangiogenesis in vivo. We show tracking of lymphatic vessel development during embryogenesis as well as lymphangiogenesis induced by specific growth factors, during wound healing and in contact hypersensitivity (CHS)--induced inflammation where we also monitor down-regulation of lymphangiogenesis by the glucocorticoid dexamethasone. Importantly, the Vegfr3-reporter allowed us to tracking tumor-induced lymphangiogenesis at the tumor periphery and in lymph nodes in association with the metastatic process. This is the first reporter mouse model for luminescence imaging of lymphangiogenesis. It should provide an important tool for studying the involvement of lymphangiogenesis in pathological processes.
Collapse
|
34
|
Zampell JC, Yan A, Avraham T, Daluvoy S, Weitman ES, Mehrara BJ. HIF-1α coordinates lymphangiogenesis during wound healing and in response to inflammation. FASEB J 2011; 26:1027-39. [PMID: 22067482 DOI: 10.1096/fj.11-195321] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This study aimed to investigate the mechanisms that coordinate lymphangiogenesis. Using mouse models of lymphatic regeneration and inflammatory lymphangiogenesis, we explored the hypothesis that hypoxia inducible factor-α (HIF-1α) is a central regulator of lymphangiogenesis. We show that HIF-1α inhibition by small molecule inhibitors (YC-1 and 2-methyoxyestradiol) results in delayed lymphatic repair, decreased local vascular endothelial growth factor-C (VEGF-C) expression, reduced numbers of VEGF-C(+) cells, and reductions in inflammatory lymphangiogenesis. Using transgenic HIF-1α/luciferase mice to image HIF-1α expression in real time in addition to Western blot analysis and pimonidazole staining for cellular hypoxia, we demonstrate that hypoxia stabilizes HIF-1α during initial stages of wound repair (1-2 wk); whereas inflammation secondary to gradients of lymphatic fluid stasis stabilizes HIF-1α thereafter (3-6 wk). In addition, we show that CD4(+) cell-mediated inflammation is necessary for this response and regulates HIF-1α expression by macrophages, as CD4-deficient or CD4-depleted mice demonstrate 2-fold reductions in HIF-1α expression as compared to wild-types. In summary, we show that HIF-1α is a critical coordinator of lymphangiogenesis by regulating the expression of lymphangiogenic cytokines as part of an early response mechanism to hypoxia, inflammation, and lymphatic fluid stasis.
Collapse
Affiliation(s)
- Jamie C Zampell
- Division of Plastic and Reconstructive Surgery and Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
35
|
Kashiwagi S, Hosono K, Suzuki T, Takeda A, Uchinuma E, Majima M. Role of COX-2 in lymphangiogenesis and restoration of lymphatic flow in secondary lymphedema. J Transl Med 2011; 91:1314-25. [PMID: 21577211 DOI: 10.1038/labinvest.2011.84] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The pathophysiology of secondary lymphedema remains poorly understood. To clarify the roles of cyclooxygenase (COX)-2 in enhancement of lymphangiogenesis during secondary lymphedema, we tested a mouse tail model and evaluated the recurrence of lymph flow. To induce lymphedema, a circumferential incision was made in the tail of anesthetized mice to sever the dermal lymphatic vessels. The maximum diameters of the tails were measured weekly. We found that the diameters of the tails around the wounds were markedly increased after surgery, and reached maximum size 2 weeks after wounding in mice without a COX-2 inhibitor, celecoxib (Celecoxib-). Expression of COX-2 in wound granulation tissues was markedly increased 1 week after surgery compared with unwounded naive control mice. In Celecoxib-, recurrence of lymphatic flow in the wound granulation tissues was detected 3 weeks after surgical treatment. In contrast, lymphatic flow was markedly suppressed in mice treated with celecoxib (Celecoxib+). Newly formed lymphatic structures were identified in the granulation tissues formed at wounded lesions in Celecoxib-, whereas those were markedly suppressed in Celecoxib+. Interstitial tissue pressures in the distal areas of the tail wounds were markedly increased in Celecoxib+ with reduced expression of vascular endothelial cell growth factor (VEGF)-C. F4/80-positive cells were accumulated to the wound granulation tissues in Celecoxib-, and the accumulation of these cells was suppressed in Celecoxib+. Prostaglandin E(2) (PGE(2)) upregulated the expressions of VEGF-A and VEGF-C in cultured macrophages, but not human lymphatic microvascular endothelial cells. The present study therefore suggests that lymphangiogenesis, together with recurrence of lymph flow after surgical induction of lymphedema, is upregulated by COX-2 possibly via generation of PGs.
Collapse
Affiliation(s)
- Shinya Kashiwagi
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Differential mRNA and tissue expression of lymphangiogenic growth factors (VEGF-C and -D) and their receptor (VEGFR-3) during tail regeneration in a gecko. J Comp Physiol B 2011; 182:109-26. [DOI: 10.1007/s00360-011-0604-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 06/28/2011] [Accepted: 07/04/2011] [Indexed: 10/17/2022]
|
37
|
Fujita G, Sato S, Kishino M, Iwai SI, Nakazawa M, Toyosawa S, Yura Y, Ogawa Y. Lymphatic vessels and related factors in adenoid cystic carcinoma of the salivary gland. Mod Pathol 2011; 24:885-91. [PMID: 21423152 DOI: 10.1038/modpathol.2011.42] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenoid cystic carcinoma of the salivary gland preferentially metastasizes to distant organs. It rarely metastasizes to lymph nodes. Recently, lymphangiogenesis has been associated with lymph node metastasis. Therefore, lymphangiogenesis in adenoid cystic carcinoma was evaluated from the number of lymphatic vessels and the expression of lymphangiogenic factors. Immunohistochemistry and molecular analysis were performed on clinical materials (29 cases for immunohistochemistry and 9 cases for molecular analysis). Normal submandibular gland was used as a negative control of lymphangiogenesis (10 cases for immunohistochemistry and 5 cases for molecular analysis). In adenoid cystic carcinoma, podoplanin-positive lymphatic vessels were small and often constricted, and localized to the tumor periphery. They did not have Ki67-positive endothelial cells. The lymphatic vessel density of the tumor did not exceed that of the salivary gland. By reverse transcriptase-polymerase chain reaction, adenoid cystic carcinoma and the salivary gland expressed vascular endothelial growth factor receptor-3 (VEGFR-3) similarly but VEGF-C and VEGF-D differently. Adenoid cystic carcinoma expressed VEGF-C, whereas the salivary gland expressed both VEGF-C and VEGF-D. VEGF-C was weak in adenoid cystic carcinoma and strong in the salivary gland. Real-time reverse transcriptase-polymerase chain reaction of VEGF-C showed that the ratio of the tumor to the salivary gland was 1 to 30 (P<0.01). Immunohistochemistry barely detected VEGF-C in adenoid cystic carcinoma. VEGF-C was expressed faintly by the tumor cells. VEGF-C and VEGF-D were detected in the serous acinar and duct cells and in the duct contents in the salivary gland. VEGFR-3 appeared to be expressed by lymphatic vessels in both adenoid cystic carcinoma and the salivary gland. These results indicate that lymphangiogenesis does not occur in adenoid cystic carcinoma. This condition would lead to the uncommon lymphatic metastasis.
Collapse
Affiliation(s)
- Gentaro Fujita
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Several gene therapy approaches have been designed for the treatment of cardiovascular diseases. A positive finding is that the safety of cardiovascular gene therapy has been excellent even in long-term follow-up. However, several hurdles to this field are still present. A major disappointing feature of the trials is that while preclinical and uncontrolled phase-I gene therapy trials have been positive, none of the randomized controlled phase-II/III cardiovascular gene therapy trials have shown clinically relevant positive effects. Low gene transfer efficiency seems to be associated with several trials. A sophisticated efficient delivery method for cardiovascular applications is still lacking and only low concentrations of the gene product are produced in the target tissues. Only a few gene therapy vectors can be produced in large scale. In addition, inflammatory reactions against vectors and inability to regulate gene expression are still present. Furthermore, a strong placebo effect is affecting the results in gene therapy trials, and long-term trials have become more difficult to conduct because of the multiplicity of therapies applied simultaneously on the patients. This review summarizes advances and obstacles of current cardiovascular clinical gene therapy trials.
Collapse
|
39
|
Affiliation(s)
- Camilla Norrmén
- From the Molecular/Cancer Biology Laboratory, Research Programs Unit, Institute for Molecular Medicine Finland and Helsinki University Hospital, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland (C.N., T.T., K.A.), and Division of Experimental Oncology and Department of Biochemistry, CePO, University of Lausanne and CHUV, Epalinges, Switzerland (T.V.P.)
| | - Tuomas Tammela
- From the Molecular/Cancer Biology Laboratory, Research Programs Unit, Institute for Molecular Medicine Finland and Helsinki University Hospital, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland (C.N., T.T., K.A.), and Division of Experimental Oncology and Department of Biochemistry, CePO, University of Lausanne and CHUV, Epalinges, Switzerland (T.V.P.)
| | - Tatiana V. Petrova
- From the Molecular/Cancer Biology Laboratory, Research Programs Unit, Institute for Molecular Medicine Finland and Helsinki University Hospital, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland (C.N., T.T., K.A.), and Division of Experimental Oncology and Department of Biochemistry, CePO, University of Lausanne and CHUV, Epalinges, Switzerland (T.V.P.)
| | - Kari Alitalo
- From the Molecular/Cancer Biology Laboratory, Research Programs Unit, Institute for Molecular Medicine Finland and Helsinki University Hospital, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland (C.N., T.T., K.A.), and Division of Experimental Oncology and Department of Biochemistry, CePO, University of Lausanne and CHUV, Epalinges, Switzerland (T.V.P.)
| |
Collapse
|
40
|
Yan A, Avraham T, Zampell JC, Aschen SZ, Mehrara BJ. Mechanisms of lymphatic regeneration after tissue transfer. PLoS One 2011; 6:e17201. [PMID: 21359148 PMCID: PMC3040774 DOI: 10.1371/journal.pone.0017201] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 01/24/2011] [Indexed: 11/19/2022] Open
Abstract
Introduction Lymphedema is the chronic swelling of an extremity that occurs commonly after lymph node resection for cancer treatment. Recent studies have demonstrated that transfer of healthy tissues can be used as a means of bypassing damaged lymphatics and ameliorating lymphedema. The purpose of these studies was to investigate the mechanisms that regulate lymphatic regeneration after tissue transfer. Methods Nude mice (recipients) underwent 2-mm tail skin excisions that were either left open or repaired with full-thickness skin grafts harvested from donor transgenic mice that expressed green fluorescent protein in all tissues or from LYVE-1 knockout mice. Lymphatic regeneration, expression of VEGF-C, macrophage infiltration, and potential for skin grafting to bypass damaged lymphatics were assessed. Results Skin grafts healed rapidly and restored lymphatic flow. Lymphatic regeneration occurred beginning at the peripheral edges of the graft, primarily from ingrowth of new lymphatic vessels originating from the recipient mouse. In addition, donor lymphatic vessels appeared to spontaneously re-anastomose with recipient vessels. Patterns of VEGF-C expression and macrophage infiltration were temporally and spatially associated with lymphatic regeneration. When compared to mice treated with excision only, there was a 4-fold decrease in tail volumes, 2.5-fold increase in lymphatic transport by lymphoscintigraphy, 40% decrease in dermal thickness, and 54% decrease in scar index in skin-grafted animals, indicating that tissue transfer could bypass damaged lymphatics and promote rapid lymphatic regeneration. Conclusions Our studies suggest that lymphatic regeneration after tissue transfer occurs by ingrowth of lymphatic vessels and spontaneous re-connection of existing lymphatics. This process is temporally and spatially associated with VEGF-C expression and macrophage infiltration. Finally, tissue transfer can be used to bypass damaged lymphatics and promote rapid lymphatic regeneration.
Collapse
Affiliation(s)
- Alan Yan
- The Division of Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | | | | | | |
Collapse
|
41
|
Lähteenvuo M, Honkonen K, Tervala T, Tammela T, Suominen E, Lähteenvuo J, Kholová I, Alitalo K, Ylä-Herttuala S, Saaristo A. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation 2011; 123:613-20. [PMID: 21282502 DOI: 10.1161/circulationaha.110.965384] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lymphedema after surgery, infection, or radiation therapy is a common and often incurable problem. Application of lymphangiogenic growth factors has been shown to induce lymphangiogenesis and to reduce tissue edema. The therapeutic effect of autologous lymph node transfer combined with adenoviral growth factor expression was evaluated in a newly established porcine model of limb lymphedema. METHODS AND RESULTS The lymphatic vasculature was destroyed within a 3-cm radius around an inguinal lymph node. Lymph node grafts and adenovirally (Ad) delivered vascular endothelial growth factor (VEGF)-C (n=5) or VEGF-D (n=9) were used to reconstruct the lymphatic network in the inguinal area; AdLacZ (β-galactosidase; n=5) served as a control. Both growth factors induced robust growth of new lymphatic vessels in the defect area, and postoperative lymphatic drainage was significantly improved in the VEGF-C/D-treated pigs compared with controls. The structure of the transferred lymph nodes was best preserved in the VEGF-C-treated pigs. Interestingly, VEGF-D transiently increased accumulation of seroma fluid in the operated inguinal region postoperatively, whereas VEGF-C did not have this side effect. CONCLUSIONS These results show that growth factor gene therapy coupled with lymph node transfer can be used to repair damaged lymphatic networks in a large animal model and provide a basis for future clinical trials of the treatment of lymphedema.
Collapse
|
42
|
Wang Y, Oliver G. Current views on the function of the lymphatic vasculature in health and disease. Genes Dev 2010; 24:2115-26. [PMID: 20889712 DOI: 10.1101/gad.1955910] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lymphatic vascular system is essential for lipid absorption, fluid homeostasis, and immune surveillance. Until recently, lymphatic vessel dysfunction had been associated with symptomatic pathologic conditions such as lymphedema. Work in the last few years had led to a better understanding of the functional roles of this vascular system in health and disease. Furthermore, recent work has also unraveled additional functional roles of the lymphatic vasculature in fat metabolism, obesity, inflammation, and the regulation of salt storage in hypertension. In this review, we summarize the functional roles of the lymphatic vasculature in health and disease.
Collapse
Affiliation(s)
- Yingdi Wang
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | |
Collapse
|
43
|
Ongstad EL, Bouta EM, Roberts JE, Uzarski JS, Gibbs SE, Sabel MS, Cimmino VM, Roberts MA, Goldman J. Lymphangiogenesis-independent resolution of experimental edema. Am J Physiol Heart Circ Physiol 2010; 299:H46-54. [PMID: 20207821 DOI: 10.1152/ajpheart.00008.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor (VEGF)-C is necessary for lymphangiogenesis, and excess VEGF-C has been shown to be ameliorative for edema produced by lymphatic obstruction in experimental models. However, it has recently been shown that edema can resolve in the mouse tail even in the complete absence of capillary lymphangiogenesis when distal lymph fluid crosses the regenerating wound site interstitially. This finding has raised questions about the action of VEGF-C/VEGF receptor (VEGFR) signaling during the resolution of experimental edema. Here, the roles of VEGFR-2 and VEGFR-3 signaling in edema resolution were explored. It was found that edema resolved following neutralization of either VEGFR-2 or VEGFR-3 in the mouse tail skin, which inhibited lymphangiogenesis. Neutralization of either VEGFR-2 or VEGFR-3 reduced angiogenesis at the site of obstruction at day 10 (9.2 +/- 1.2% and 11.5 +/- 1.0% blood capillary coverage, respectively) relative to controls (14.3 +/- 1.5% blood capillary coverage). Combined VEGFR-2/-3 neutralization more strongly inhibited angiogenesis (6.9 +/- 1.5% blood capillary coverage), leading to a reduced wound repair of the lymphatic obstruction and extended edema in the tail skin. In contrast, improved tissue repair of the obstruction site increased edema resolution. Macrophages in the swollen tissue were excluded as contributing factors in the VEGFR-dependent extended edema. These results support a role for VEGFR-2/-3-combined signaling in the resolution of experimental edema that is lymphangiogenesis independent.
Collapse
Affiliation(s)
- Emily L Ongstad
- Biomedical Engineering Department, Michigan Technological University, Houghton, MI 49931, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Koch M, Dettori D, Van Nuffelen A, Souffreau J, Marconcini L, Wallays G, Moons L, Bruyère F, Oliviero S, Noel A, Foidart JM, Carmeliet P, Dewerchin M. VEGF-D deficiency in mice does not affect embryonic or postnatal lymphangiogenesis but reduces lymphatic metastasis. J Pathol 2010; 219:356-64. [PMID: 19718705 DOI: 10.1002/path.2605] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factor-D (VEGF-D) is one of the two ligands of the VEGFR-3 receptor on lymphatic endothelial cells. Gene-silencing studies in mice and Xenopus tadpoles recently showed that the role of endogenous VEGF-D in lymphatic development is moderate. By contrast, exogenous VEGF-D is capable of stimulating lymphangiogenesis. Nonetheless, its endogenous role in pathological conditions remains largely unknown. Hence, we reassessed its role in disease, using Vegf-d(null) mice. Vegf-d(null) mice were generated that, under physiological conditions, displayed normal embryonic and postnatal lymphangiogenesis and lymphatic remodelling, efficient lymphatic functioning and normal health. Vegf-d(null) mice also reponded normally in models of skin wound healing and healing of infarcted myocardium, despite enhanced expression of VEGF-D in these models in wild-type mice. In contrast, Vegf-d(null) mice displayed reduced peritumoral lymphangiogenesis and lymph node metastasis in an orthotopic pancreatic tumour model. Together, our data indicate that endogenous VEGF-D in mice is dispensible for lymphangiogenesis during development, in postnatal and adult physiology and in several pathological conditions, but significantly contributes to lymphatic metastasis.
Collapse
Affiliation(s)
- Marta Koch
- Vesalius Research Centre, VIB, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kajiya K, Sawane M, Huggenberger R, Detmar M. Activation of the VEGFR-3 Pathway by VEGF-C Attenuates UVB-Induced Edema Formation and Skin Inflammation by Promoting Lymphangiogenesis. J Invest Dermatol 2009; 129:1292-8. [DOI: 10.1038/jid.2008.351] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
Critical role of CD11b+ macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution. Blood 2009; 113:5650-9. [PMID: 19346498 DOI: 10.1182/blood-2008-09-176776] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Using a bacterial pathogen-induced acute inflammation model in the skin, we defined the roles of local lymphatic vessels and draining lymph nodes (DLNs) in antigen clearance and inflammation resolution. At the peak day of inflammation, robust expansion of lymphatic vessels and profound infiltration of CD11b+/Gr-1+ macrophages into the inflamed skin and DLN were observed. Moreover, lymph flow and inflammatory cell migration from the inflamed skin to DLNs were enhanced. Concomitantly, the expression of lymphangiogenic growth factors such as vascular endothelial growth factor C (VEGF-C), VEGF-D, and VEGF-A were significantly up-regulated in the inflamed skin, DLNs, and particularly in enriched CD11b+ macrophages from the DLNs. Depletion of macrophages, or blockade of VEGF-C/D or VEGF-A, largely attenuated these phenomena, and produced notably delayed antigen clearance and inflammation resolution. Conversely, keratin 14 (K14)-VEGF-C transgenic mice, which have dense and enlarged lymphatic vessels in the skin dermis, exhibited accelerated migration of inflammatory cells from the inflamed skin to the DLNs and faster antigen clearance and inflammation resolution. Taken together, these results indicate that VEGF-C, -D, and -A derived from the CD11b+/Gr-1+ macrophages and local inflamed tissues play a critical role in promoting antigen clearance and inflammation resolution.
Collapse
|
47
|
Lohela M, Bry M, Tammela T, Alitalo K. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol 2009; 21:154-65. [DOI: 10.1016/j.ceb.2008.12.012] [Citation(s) in RCA: 543] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 12/30/2008] [Indexed: 01/01/2023]
|
48
|
Dunworth WP, Caron KM. G protein-coupled receptors as potential drug targets for lymphangiogenesis and lymphatic vascular diseases. Arterioscler Thromb Vasc Biol 2009; 29:650-6. [PMID: 19265032 DOI: 10.1161/atvbaha.109.185066] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are widely expressed cell surface receptors that have been successfully exploited for the treatment of a variety of human diseases. Recent studies in genetically engineered mouse models have led to the identification of several GPCRs important for lymphatic vascular development and function. The adrenomedullin receptor, which consists of an oligomer between calcitonin receptor-like receptor and receptor activity modifying protein 2, is required for normal lymphatic vascular development and regulates lymphatic capillary permeability in mice. Numerous studies also suggest that lysophospholipid receptors are involved in the development of lymphatic vessels and lymphatic endothelial cell permeability. Given our current lack of pharmacological targets for the treatment of lymphatic vascular diseases like lymphedema, the continued identification and study of GPCRs in lymphatic endothelial cells may eventually lead to major breakthroughs and new pharmacological strategies for the treatment of lymphedema.
Collapse
Affiliation(s)
- William P Dunworth
- Department of Cell and Molecular Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
49
|
Stanton AW, Modi S, Mellor RH, Levick JR, Mortimer PS. Recent Advances in Breast Cancer-Related Lymphedema of the Arm: Lymphatic Pump Failure and Predisposing Factors. Lymphat Res Biol 2009; 7:29-45. [PMID: 19302022 DOI: 10.1089/lrb.2008.1026] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Anthony W.B. Stanton
- Divisions of Cardiac & Vascular Sciences (Dermatology), St George's Hospital Medical School, University of London, United Kingdom
| | - Stephanie Modi
- Divisions of Cardiac & Vascular Sciences (Dermatology), St George's Hospital Medical School, University of London, United Kingdom
| | - Russell H. Mellor
- Divisions of Cardiac & Vascular Sciences (Dermatology), St George's Hospital Medical School, University of London, United Kingdom
| | - J. Rodney Levick
- Basic Medical Sciences (Physiology), St George's Hospital Medical School, University of London, United Kingdom
| | - Peter S. Mortimer
- Divisions of Cardiac & Vascular Sciences (Dermatology), St George's Hospital Medical School, University of London, United Kingdom
| |
Collapse
|
50
|
Jin DP, An A, Liu J, Nakamura K, Rockson SG. Therapeutic Responses to Exogenous VEGF-C Administration in Experimental Lymphedema: Immunohistochemical and Molecular Characterization. Lymphat Res Biol 2009; 7:47-57. [DOI: 10.1089/lrb.2009.0002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Da Pan Jin
- Stanford University School of Medicine, Stanford, California
| | - Andrew An
- Stanford University School of Medicine, Stanford, California
| | - Joseph Liu
- Stanford University School of Medicine, Stanford, California
| | - Kenta Nakamura
- Stanford University School of Medicine, Stanford, California
| | | |
Collapse
|