1
|
Campos JL, Suominen S, Pons G, Al-Sakkaf AM, Lusetti IL, Sirota M, Vela FJ, Pires L, Sánchez-Margallo FM, Abellán E, Masiá J. Lymphatic Patterns in the Superficial Circumflex Iliac Artery Perforator Flap. J Reconstr Microsurg 2024. [PMID: 38848754 DOI: 10.1055/a-2340-9629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
BACKGROUND Lymphedema is a chronic condition, characterized by fluid buildup and tissue swelling and is caused by impairment of the lymphatic system. The lymph interpositional flap transfer technique, in which lymph flow is restored with a flap that includes subdermal lymphatic channels, is an option for surgical reconstruction. The superficial circumflex iliac artery perforator (SCIP) flap can be used for this purpose. This study aimed to describe and characterize the lymphatic patterns within the vascular territory of the SCIP flap. METHODS This cross-sectional multicenter study involved 19 healthy volunteers aged ≥18 years of both sexes assessing the bilateral SCIP flap zone. Superficial lymphatic patterns were evaluated at 4-, 14-, and 24 minutes after indocyanine green (ICG) lymphography injection. Standardized procedures were implemented for all participants in both hospitals. RESULTS The linear pattern was predominant bilaterally. The median number of lymphatic vessels and their length increased over time. Most lymphatic vessels in the SCIP flap were oriented toward the inguinal lymph node (ILN). However, the left SCIP zone lymphatic vessels were directed opposite to the ILN. CONCLUSION The two sides SCIP zones were not significantly different. The primary direction of the bilateral lymphatic vessels was toward the ILN, although only single-side lymphatic vessels were in the opposite direction. These findings emphasize the importance of assessing lymphatic axiality and coherent lymphatic patterns prior to undertaking the SCIP as an interposition flap, to ensure effective restoration of lymphatic flow.
Collapse
Affiliation(s)
- José Luis Campos
- Department of Microsurgery, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Sinikka Suominen
- Department of Plastic and Reconstructive Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Gemma Pons
- Department of Plastic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ali M Al-Sakkaf
- Department of Plastic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Irene Laura Lusetti
- Department of Plastic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Max Sirota
- Department of Plastic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Javier Vela
- Department of Microsurgery, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Laura Pires
- Department of Microsurgery, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | | | - Elena Abellán
- Department of Microsurgery, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Jaume Masiá
- Department of Plastic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
2
|
Zhang Y, Wu Z, Zhao Q, Liu Y, Huang Q, Zhang M, Li S, Wang D, Li N, Chi Y, Liu Y. Mesenteric Lymphatic B Cells Migrate to the Intestine and Aggravate DSS-Induced Colitis via the CXCR5-CXCL13 Axis. BIOLOGY 2024; 13:322. [PMID: 38785804 PMCID: PMC11117591 DOI: 10.3390/biology13050322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is still unknown. Mesenteric lymphatics (MLs), which are closely related to the intestine in both anatomy and physiology, have been suggested to be involved in IBD. In the present study, we aim to investigate the effects of ML immune cells on IBD and explore the potential associated mechanisms. Acute colitis was induced in rats using dextran sulfate sodium salt (DSS). Mesenteric lymphangiogenesis, ML stenosis, and dilation were observed, with an increased proportion of MLB cells in DSS-induced colitis rats. The adoptive transfer of B cells isolated from ML (MLB) was employed to investigate their effects on colitis. MLB cells derived from DSS-induced colitis rats exhibited a higher propensity to migrate to the intestine. The proportion of colonic T cells was altered, along with the aggravated colitis induced by the adoptive transfer of MLB cells derived from DSS-induced colitis rats. RNA sequencing revealed increased Cxcr5 expression in MLB cells from colitis rats, while real-time PCR indicated an upregulation of its ligand Cxcl13 in the colon of colitis rats. These findings suggest that MLB cells may migrate to the intestine and aggravate colitis. In summary, colonic T cells respond to MLB cells from colitis rats, and MLB cells aggravate DSS-induced colitis via the CXCR5-CXCL13 axis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Zhe Wu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Qinghe Zhao
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yaming Liu
- Department of Gastroenterology and Hepatology, Xiamen University Zhongshan Hospital, Xiamen 361001, China;
| | - Qing Huang
- Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China;
| | - Menglei Zhang
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Shuolei Li
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Na Li
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yujing Chi
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
3
|
Bhatnagar M, Fisher A, Ramsaroop S, Carter A, Pippard B. Chylothorax: pathophysiology, diagnosis, and management-a comprehensive review. J Thorac Dis 2024; 16:1645-1661. [PMID: 38505027 PMCID: PMC10944732 DOI: 10.21037/jtd-23-1636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024]
Abstract
Chylothorax is a rare condition characterized by the accumulation of chyle in the pleural space. While it accounts for a small percentage of pleural effusions, chylothorax can lead to significant morbidity and mortality. This article provides a comprehensive overview of chylothorax, covering its relevant anatomy, aetiology, pathophysiology, clinical features, diagnosis, and management. Injury or disruption to the thoracic duct (which is responsible for chyle transport) leads to the development of chylothorax. This may result from trauma, such as iatrogenic injury during surgery, or non-traumatic causes, including malignancy, lymphatic disorders, and heart failure. Recognition of the underlying cause is essential to tailor management. Clinical presentation varies, with symptoms linked to rate of chyle accumulation and the causative condition. Diagnosis relies on pleural fluid analysis, with demonstration of elevated triglyceride levels (>110 mg/dL) and reduced cholesterol levels (<200 mg/dL) being the key diagnostic criteria employed in clinical practice. Various imaging modalities, including computed tomography (CT) scans and lymphatic-specific investigations, may be utilised to aid identification of the site of chyle leak, as well as determine the likely underlying cause. Chylothorax management is multifaceted, with conservative approaches such as dietary modification and pharmacological interventions often initiated as first-line treatment. Drainage of chylous effusion may be necessary for symptom relief. When conservative methods fail, interventional procedures like thoracic duct ligation or embolization can be considered. Due to the diverse aetiological factors and patient characteristics associated with chylothorax, individualized management strategies are recommended. Nonetheless, management of chylothorax is an evolving field with a paucity of high-quality evidence or standardized guidelines, highlighting the importance of ongoing research and a multidisciplinary approach to optimize individual patient care.
Collapse
Affiliation(s)
- Malvika Bhatnagar
- Department of Respiratory Medicine, South Tyneside and Sunderland NHS Foundation Trust, South Tyneside, UK
| | - Annette Fisher
- Department of Respiratory Medicine, Northumbria Healthcare NHS Foundation Trust, Cramlington, UK
| | - Sudarshan Ramsaroop
- Department of Respiratory Medicine, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, UK
| | - Alison Carter
- Department of Respiratory Medicine, County Durham and Darlington NHS Foundation Trust, Durham, UK
| | - Benjamin Pippard
- Department of Respiratory Medicine, South Tyneside and Sunderland NHS Foundation Trust, South Tyneside, UK
| |
Collapse
|
4
|
Ellebrecht DB, Hoge M, von Weihe S. [Chylothorax]. Zentralbl Chir 2024; 149:133-147. [PMID: 38442889 DOI: 10.1055/a-1990-4896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Chylothorax is a serious and potentially life-threatening condition of diverse etiology. This article provides a detailed overview of anatomy, physiology, etiology, diagnosis, and therapeutic options in the context of chylothorax.
Collapse
|
5
|
Roth-Walter F, Berni Canani R, O'Mahony L, Peroni D, Sokolowska M, Vassilopoulou E, Venter C. Nutrition in chronic inflammatory conditions: Bypassing the mucosal block for micronutrients. Allergy 2024; 79:353-383. [PMID: 38084827 DOI: 10.1111/all.15972] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Nutritional Immunity is one of the most ancient innate immune responses, during which the body can restrict nutrients availability to pathogens and restricts their uptake by the gut mucosa (mucosal block). Though this can be a beneficial strategy during infection, it also is associated with non-communicable diseases-where the pathogen is missing; leading to increased morbidity and mortality as micronutritional uptake and distribution in the body is hindered. Here, we discuss the acute immune response in respect to nutrients, the opposing nutritional demands of regulatory and inflammatory cells and particularly focus on some nutrients linked with inflammation such as iron, vitamins A, Bs, C, and other antioxidants. We propose that while the absorption of certain micronutrients is hindered during inflammation, the dietary lymph path remains available. As such, several clinical trials investigated the role of the lymphatic system during protein absorption, following a ketogenic diet and an increased intake of antioxidants, vitamins, and minerals, in reducing inflammation and ameliorating disease.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Liam O'Mahony
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Diego Peroni
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Vassilopoulou
- Pediatric Area, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carina Venter
- Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
6
|
Kim S, Chun SH, Cheon YH, Kim M, Kim HO, Lee H, Hong ST, Park SJ, Park MS, Suh YS, Lee SI. Peptoniphilus gorbachii alleviates collagen-induced arthritis in mice by improving intestinal homeostasis and immune regulation. Front Immunol 2024; 14:1286387. [PMID: 38239365 PMCID: PMC10794505 DOI: 10.3389/fimmu.2023.1286387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction The intricate connection between gut microbiota and rheumatoid arthritis (RA) pathogenesis has gained prominence, although the specific microbial species contributing to RA development remain largely unknown. Recent studies have sought to comprehensively explore alterations in the human microbiome, focusing on identifying disease-related microbial species through blood analysis. Consequently, this study aimed to identify RA-associated microbial species using a serum microbial array system and to investigate the efficacy and underlying mechanisms of potential microbial species for RA treatment. Methods Serum immunoglobulin M levels against 384 intestinal microbial species were assessed using a microbial microarray in patients with RA and healthy individuals. We investigated the therapeutic potential of the identified microbial candidate regarding arthritis development, immune responses, gut barrier function, and gut microbiome using a collagen-induced arthritis (CIA) mouse model. Results Our findings revealed significant alterations in antibody levels against 36 microbial species in patients with RA compared to healthy individuals. Notably, the antibody levels against Peptoniphilus gorbachii (PG) were decreased in patients with RA and exhibited an inverse correlation with RA disease activity. In vitro experiments demonstrated that PG produced acetate and butyrate, while exhibiting anti-inflammatory properties. In CIA mice, PG administration suppressed arthritis symptoms, reduced the accumulation of inflammatory monocytes in the mesenteric lymph nodes, and downregulated gene expression of pro-inflammatory cytokines in the ileum. Additionally, PG supplementation restored intestinal barrier integrity and partially resolved gut microbial dysbiosis in CIA mice. The fecal microbiota in PG-treated mice corresponded to improved intestinal barrier integrity and reduced inflammatory responses. Conclusion This study highlights the potential of serum-based detection of anti-microbial antibodies to identify microbial targets at the species level for RA treatment. Moreover, our findings suggest that PG, identified through the microbial microarray analysis, holds therapeutic potential for RA by restoring intestinal barrier integrity and suppressing the immunologic response associated with RA.
Collapse
Affiliation(s)
- Suhee Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Sung Hak Chun
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Yun-Hong Cheon
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Mingyo Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Hyun-Ok Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Hanna Lee
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sang-Jun Park
- Research Center, BIFIDO Co, Ltd, Hongcheon, Kangwon, Republic of Korea
| | - Myeong Soo Park
- Research Center, BIFIDO Co, Ltd, Hongcheon, Kangwon, Republic of Korea
| | - Young Sun Suh
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Sang-Il Lee
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| |
Collapse
|
7
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang JL, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal Lymphatic Sphingosine-1-Phosphate Signaling Aggravates Lymphatic Dysfunction and Tissue Inflammation. Circulation 2023; 148:1231-1249. [PMID: 37609838 PMCID: PMC10592179 DOI: 10.1161/circulationaha.123.064181] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T-cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T-cell activation. Characterizing this biology is relevant for developing much needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1-deficient (S1pr1LECKO) mice were generated. Disease progression was quantified by tail-volumetric and -histopathologic measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then cocultured with CD4 T cells, followed by an analysis of CD4 T-cell activation and pathway signaling. Last, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T-cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1P receptor 1 (S1PR1). LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T-cell infiltration in mouse lymphedema. LECs, isolated from S1pr1LECKO mice and cocultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs promoted T-helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. Human dermal LECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro, P-selectin blockade reduced the activation and differentiation of Th cells cocultured with shS1PR1-treated human dermal LECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSIONS This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T-cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Lon Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Jung YJ, Lee Y, Kwon H, Kim HP, Kwon HS, Park E, Lee J, Kim YH, Maeng YS, Kwon JY. Decidual lymphatic endothelial cell-derived granulocyte-macrophage colony-stimulating factor induces M1 macrophage polarization via the NF-κB pathway in severe pre-eclampsia. Am J Reprod Immunol 2023; 90:e13744. [PMID: 37491916 DOI: 10.1111/aji.13744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/18/2023] [Accepted: 06/16/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Direct interactions between macrophages and lymphatic vessels have been shown previously. In pre-eclampsia (PE), macrophages are dominantly polarized into a proinflammatory M1 phenotype and lymphangiogenesis is defective in the decidua. Here, we investigated whether decidual lymphatic endothelial cells (dLECs) affect macrophage polarization in PE. METHOD OF STUDY THP-1 macrophages were cocultured with dLECs or cultured in the conditioned medium (CM) of dLECs. Macrophage polarization was measured using flow cytometry. Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in dLECs was measured using qRT-PCR and ELISA. The activation of nuclear translocation of nuclear factor-κ (NF-κB), an upstream signaling molecule of GM-CSF, was assessed by immunocytochemical localization of p65. Through GM-CSF knockdown and NF-κB inhibition in dLEC, we evaluated whether the GM-CSF/NF-κB pathway of PE dLEC affects decidual macrophage polarization. RESULTS The ratio of inflammatory M1 macrophages with HLA-DR+ /CD80+ markers significantly increased following coculturing with PE dLECs or culturing in PE dLEC CM, indicating that the PE dLEC-derived soluble factor acts in a paracrine manner. GM-CSF expression was significantly upregulated in PE dLECs. Recombinant human GM-CSF induced macrophage polarization toward an M1-like phenotype, whereas its knockdown in PE dLECs suppressed it, suggesting PE dLECs induce M1 macrophage polarization by secreting GM-CSF. The NF-κB p65 significantly increased in PE dLECs compared to the control, and pretreatment with an NF-κB inhibitor significantly suppressed GM-CSF production from PE dLECs. CONCLUSIONS In PE, dLECs expressing high levels of GM-CSF via the NF-κB-dependent pathway play a role in inducing decidual M1 macrophage polarization.
Collapse
Affiliation(s)
- Yun Ji Jung
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Yeji Lee
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Hayan Kwon
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, The Republic of Korea
| | - Han-Sung Kwon
- Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, The Republic of Korea
| | - Eunhyang Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, The Republic of Korea
| | - JoonHo Lee
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Young-Han Kim
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| | - Ja-Young Kwon
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Placenta-derived Stem Cell and Genomic Research Lab, Yonsei University College of Medicine, Yonsei University Health System, Seoul, The Republic of Korea
| |
Collapse
|
9
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang J, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal lymphatic S1P signaling aggravates lymphatic dysfunction and tissue inflammation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.08.23291175. [PMID: 37398237 PMCID: PMC10312855 DOI: 10.1101/2023.06.08.23291175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T cell activation. Characterizing this biology is relevant for developing much-needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1 -deficient ( S1pr1 LECKO ) mice were generated. Disease progression was quantified by tail-volumetric and -histopathological measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then co-cultured with CD4 T cells, followed by an analysis of CD4 T cell activation and pathway signaling. Finally, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1PR1. LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T cell infiltration in mouse lymphedema. LECs, isolated from S1pr1 LECKO mice and co-cultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs (HDLECs) promoted T helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. HDLECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro , P-selectin blockade reduced the activation and differentiation of Th cells co-cultured with sh S1PR1 -treated HDLECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSION This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition. Clinical Perspective What is New?: Lymphatic-specific S1pr1 deletion exacerbates lymphatic vessel malfunction and Th1/Th2 immune responses during lymphedema pathogenesis. S1pr1 -deficient LECs directly induce Th1/Th2 cell differentiation and decrease anti-inflammatory Treg populations. Peripheral dermal LECs affect CD4 T cell immune responses through direct cell contact.LEC P-selectin, regulated by S1PR1 signaling, affects CD4 T cell activation and differentiation.P-selectin blockade improves lymphedema tail swelling and decreases Th1/Th2 population in the diseased skin.What Are the Clinical Implications?: S1P/S1PR1 signaling in LECs regulates inflammation in lymphedema tissue.S1PR1 expression levels on LECs may be a useful biomarker for assessing predisposition to lymphatic disease, such as at-risk women undergoing mastectomyP-selectin Inhibitors may be effective for certain forms of lymphedema.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
10
|
Baker ML, Cantley LG. The Lymphatic System in Kidney Disease. KIDNEY360 2023; 4:e841-e850. [PMID: 37019177 PMCID: PMC10371377 DOI: 10.34067/kid.0000000000000120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 04/07/2023]
Abstract
The high-capacity vessels of the lymphatic system drain extravasated fluid and macromolecules from nearly every part of the body. However, far from merely a passive conduit for fluid removal, the lymphatic system also plays a critical and active role in immune surveillance and immune response modulation through the presentation of fluid, macromolecules, and trafficking immune cells to surveillance cells in regional draining lymph nodes before their return to the systemic circulation. The potential effect of this system in numerous disease states both within and outside of the kidney is increasingly being explored for their therapeutic potential. In the kidneys, the lymphatics play a critical role in both fluid and macromolecule removal to maintain oncotic and hydrostatic pressure gradients for normal kidney function, as well as in shaping kidney immunity, and potentially in balancing physiological pathways that promote healthy organ maintenance and responses to injury. In many states of kidney disease, including AKI, the demand on the preexisting lymphatic network increases for clearance of injury-related tissue edema and inflammatory infiltrates. Lymphangiogenesis, stimulated by macrophages, injured resident cells, and other drivers in kidney tissue, is highly prevalent in settings of AKI, CKD, and transplantation. Accumulating evidence points toward lymphangiogenesis being possibly harmful in AKI and kidney allograft rejection, which would potentially position lymphatics as another target for novel therapies to improve outcomes. However, the extent to which lymphangiogenesis is protective rather than maladaptive in the kidney in various settings remains poorly understood and thus an area of active research.
Collapse
Affiliation(s)
- Megan L Baker
- Section of Nephrology, Yale School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
11
|
Trivedi A, Reed HO. The lymphatic vasculature in lung function and respiratory disease. Front Med (Lausanne) 2023; 10:1118583. [PMID: 36999077 PMCID: PMC10043242 DOI: 10.3389/fmed.2023.1118583] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
The lymphatic vasculature maintains tissue homeostasis via fluid drainage in the form of lymph and immune surveillance due to migration of leukocytes through the lymphatics to the draining lymph nodes. Lymphatic endothelial cells (LECs) form the lymphatic vessels and lymph node sinuses and are key players in shaping immune responses and tolerance. In the healthy lung, the vast majority of lymphatic vessels are found along the bronchovascular structures, in the interlobular septa, and in the subpleural space. Previous studies in both mice and humans have shown that the lymphatics are necessary for lung function from the neonatal period through adulthood. Furthermore, changes in the lymphatic vasculature are observed in nearly all respiratory diseases in which they have been analyzed. Recent work has pointed to a causative role for lymphatic dysfunction in the initiation and progression of lung disease, indicating that these vessels may be active players in pathologic processes in the lung. However, the mechanisms by which defects in lung lymphatic function are pathogenic are understudied, leaving many unanswered questions. A more comprehensive understanding of the mechanistic role of morphological, functional, and molecular changes in the lung lymphatic endothelium in respiratory diseases is a promising area of research that is likely to lead to novel therapeutic targets. In this review, we will discuss our current knowledge of the structure and function of the lung lymphatics and the role of these vessels in lung homeostasis and respiratory disease.
Collapse
Affiliation(s)
- Anjali Trivedi
- Weill Cornell Medical Center, New York, NY, United States
| | - Hasina Outtz Reed
- Weill Cornell Medical Center, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Hasina Outtz Reed,
| |
Collapse
|
12
|
Ylä-Herttuala E, Vuorio T, Kettunen S, Laidinen S, Ylä-Herttuala S, Liimatainen T. Lymphatic insufficiency leads to distinct myocardial infarct content assessed by magnetic resonance T RAFFn, T 1ρ and T 2 relaxation times. Sci Rep 2023; 13:1579. [PMID: 36709358 PMCID: PMC9884273 DOI: 10.1038/s41598-023-28219-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 01/16/2023] [Indexed: 01/30/2023] Open
Abstract
The role of cardiac lymphatics in the pathogenesis of myocardial infarction (MI) is unclear. Lymphatic system regulates cardiac physiological processes such as edema and tissue fluid balance, which affect MI pathogenesis. Recently, MI and fibrosis have been assessed using endogenous contrast in magnetic resonance imaging (MRI) based on the relaxation along a fictitious field with rank n (RAFFn). We extended the RAFFn applications to evaluate the effects of lymphatic insufficiency on MI with comparison to longitudinal rotating frame (T1ρ) and T2 relaxation times. MI was induced in transgenic (TG) mice expressing soluble decoy VEGF receptor 3 that reduces lymphatic vessel formation and their wild-type (WT) control littermates for comparison. The RAFFn relaxation times with rank 2 (TRAFF2), and rank 4 (TRAFF4), T1ρ and T2 were acquired at time points 0, 3, 7, 21 and 42 days after the MI at 9.4 T. Infarct sizes were determined based on TRAFF2, TRAFF4, T1ρ and T2 relaxation time maps. The area of differences (AOD) was calculated based on the MI areas determined on T2 and TRAFF2, TRAFF4 or T1ρ relaxation time maps. Hematoxylin-eosin and Sirius red stained histology sections were prepared to confirm MI locations and sizes. MI was detected as increased TRAFF2, TRAFF4, T1ρ and T2 relaxation times. Infarct sizes were similar on all relaxation time maps during the experimental period. Significantly larger AOD values were found together with increased AOD values in the TG group compared to the WT group. Histology confirmed these findings. The lymphatic deficiency was found to increase cardiac edema in MI. The combination of TRAFF2 (or TRAFF4) and T2 characterizes MI and edema in the myocardium in both lymphatic insufficiency and normal mice without any contrast agents.
Collapse
Affiliation(s)
- Elias Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Clinical Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Taina Vuorio
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanna Kettunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Svetlana Laidinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Timo Liimatainen
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland. .,Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
13
|
Sakurai Y, Yoshikawa K, Arai K, Kazaoka A, Aoki S, Ito K, Nakai Y, Tange K, Furihata T, Tanaka H, Akita H. siRNA delivery to lymphatic endothelial cells via ApoE-mediated uptake by lipid nanoparticles. J Control Release 2023; 353:125-133. [PMID: 36414194 DOI: 10.1016/j.jconrel.2022.11.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/27/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022]
Abstract
Systemically administered lipid nanoparticles (LNPs) are complexed with Apolipoprotein E (ApoE) in the bloodstream, and the complex is subsequently largely taken up by hepatocytes. Based on a previous report showing that, like blood, lymph fluid also contains ApoE, and that LECs, in turn, expresses a low density-lipoprotein receptor (LDLR), which is the receptor responsible for the ApoE-bound LNP, we hypothesized that subcutaneously administered LNPs would be taken up by LECs via an ApoE-LDLR pathway. Our in vitro studies using immortal LECs that we established in a previous study showed that LEC indeed took up LNPs in an ApoE-dependent manner. We then reported on the development of LNPs that target the lymphatic endothelium for in vivo siRNA delivery after subcutaneous administration. The key to success for in vivo LEC targeting is that the surface needs to be modified with a high density of polyethylene glycol (PEG)-conjugated lipids with short acyl chains (C14). The LNPs were drained into the lymphatic system, and then accumulated in lymphatic endothelial cells in an ApoE-dependent manner, most likely after the release of the PEG-lipid. Subcutaneous administration of optimized LNPs containing encapsulated siRNA against VEGFR3, a marker of LECs, significantly inhibited the expression of VEGFR3. These findings are the first report of a simple straightforward strategy for targeting lymphatic endothelial cells by using ionizable lipid-formulated LNPs.
Collapse
Affiliation(s)
- Yu Sakurai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Keito Yoshikawa
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Kenta Arai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Akira Kazaoka
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Shigeki Aoki
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yuta Nakai
- DDS Research Laboratory, NOF Corporation, Kanagawa 210-0865, Japan
| | - Kota Tange
- DDS Research Laboratory, NOF Corporation, Kanagawa 210-0865, Japan
| | - Tomomi Furihata
- Laboratory of Clinical Pharmacy & Experimental Therapeutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
14
|
Sedbon T, Azuelos A, Bosc R, D’Andrea F, Pensato R, Maruccia M, Meningaud JP, Hersant B, La Padula S. Spontaneous Lymph Flow Restoration in Free Flaps: A Pilot Study. J Clin Med 2022; 12:jcm12010229. [PMID: 36615028 PMCID: PMC9821224 DOI: 10.3390/jcm12010229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Oncologic excision and trauma can be responsible for major defects and lymphedema. Free flaps are commonly used for reconstruction. We aimed to determine if lymphatic flow between flap and recipient site can be restored without lymphatic surgery. METHODS 15 free flaps were performed in different patients in our center. Infrared-based lymphography was used to plan surgery. Indocyanine green (ICG) was injected in the flap's subdermal tissue and also at the edges of the skin defect. Circumferential lymphatic channels were marked 5 min after the ICG injection. Fluorescent images were recorded with an infrared camera system. The flap inset was obtained by putting side to side the flap markings and the recipient site markings. Infrared-based lymphography was performed on every patient one year after surgery. Spontaneous lymph flow restoration was judged positive if lymphatic connections were observed between the flap and the recipient site. RESULTS seven free ALT and eight DIEP flaps were performed. All ALT flaps were designed following the limb axis which is the lymphatic axiality. Spontaneous lymph flow restoration was observed for the seven ALT flaps. Eight DIEP flaps were designed upside down and one was designed following the lymph axiality. Spontaneous lymph flow restoration was only observed for the one designed following the lymph axiality. CONCLUSIONS designing reconstructive free flap regarding lymph axiality seems to improve spontaneous lymph flow restoration between flap and recipient site without any specific lymphatic surgery.
Collapse
Affiliation(s)
- Théo Sedbon
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Arié Azuelos
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Romain Bosc
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Francesco D’Andrea
- Department of Plastic and Reconstructive Surgery, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Rosita Pensato
- Department of Plastic and Reconstructive Surgery, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Michele Maruccia
- Unit of Plastic, Reconstructive Surgery and Burn Center, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Jean Paul Meningaud
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Barbara Hersant
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
| | - Simone La Padula
- Department of Plastic, Reconstructive and Maxillo Facial Surgery, Henri Mondor Hospital, University Paris XII, 51 Avenue du Maréchal de Lattre de Tassigny, 94000 Créteil, France
- Department of Plastic and Reconstructive Surgery, Università Degli Studi di Napoli Federico II, Via Pansini 5, 80131 Napoli, Italy
- Correspondence:
| |
Collapse
|
15
|
The Impact of Stem/Progenitor Cells on Lymphangiogenesis in Vascular Disease. Cells 2022; 11:cells11244056. [PMID: 36552820 PMCID: PMC9776475 DOI: 10.3390/cells11244056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022] Open
Abstract
Lymphatic vessels, as the main tube network of fluid drainage and leukocyte transfer, are responsible for the maintenance of homeostasis and pathological repairment. Recently, by using genetic lineage tracing and single-cell RNA sequencing techniques, significant cognitive progress has been made about the impact of stem/progenitor cells during lymphangiogenesis. In the embryonic stage, the lymphatic network is primarily formed through self-proliferation and polarized-sprouting from the lymph sacs. However, the assembly of lymphatic stem/progenitor cells also guarantees the sustained growth of lymphvasculogenesis to obtain the entire function. In addition, there are abundant sources of stem/progenitor cells in postnatal tissues, including circulating progenitors, mesenchymal stem cells, and adipose tissue stem cells, which can directly differentiate into lymphatic endothelial cells and participate in lymphangiogenesis. Specifically, recent reports indicated a novel function of lymphangiogenesis in transplant arteriosclerosis and atherosclerosis. In the present review, we summarized the latest evidence about the diversity and incorporation of stem/progenitor cells in lymphatic vasculature during both the embryonic and postnatal stages, with emphasis on the impact of lymphangiogenesis in the development of vascular diseases to provide a rational guidance for future research.
Collapse
|
16
|
Nurlaila I, Roh K, Yeom CH, Kang H, Lee S. Acquired lymphedema: Molecular contributors and future directions for developing intervention strategies. Front Pharmacol 2022; 13:873650. [PMID: 36386144 PMCID: PMC9640931 DOI: 10.3389/fphar.2022.873650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/13/2022] [Indexed: 08/05/2023] Open
Abstract
Lymphedema is a debilitating chronic disease that mostly develops as an adverse reaction to cancer treatment modalities such as chemotherapy, surgery, and radiotherapy. Lymphedema also appears to be a deteriorating consequence of roundworm infections, as best represented by filariasis. According to its origin, lymphedema is classified as primary lymphedema and acquired lymphedema. The latter is an acquired condition that, hitherto, received a considerably low attention owing to the less number of fatal cases been reported. Notably, despite the low mortality rate in lymphedema, it has been widely reported to reduce the disease-free survival and thus the quality of life of affected patients. Hence, in this review, we focused on acquired lymphedema and orchestration of molecular interplays associated with either stimulation or inhibition of lymphedema development that were, in vast majority, clearly depicted in animal models with their specific and distinct technical approaches. We also discussed some recent progress made in phytochemical-based anti-lymphedema intervention strategies and the specific mechanisms underlying their anti-lymphedema properties. This review is crucial to understand not only the comprehensive aspects of the disease but also the future directions of the intervention strategies that can address the quality of life of affected patients rather than alleviating apparent symptoms only.
Collapse
Affiliation(s)
- Ika Nurlaila
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
- Department of Vaccine and Drugs, The National Research and Innovation Agency, Jakarta, Indonesia
| | - Kangsan Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Division of Cardiology and Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Hee Kang
- Humanitas College, Kyung Hee University, Yongin, South Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
17
|
Liu Y, Chen Q, Ren R, Zhang Q, Yan G, Yin D, Zhang M, Yang Y. Platycodon grandiflorus polysaccharides deeply participate in the anti-chronic bronchitis effects of platycodon grandiflorus decoction, a representative of “the lung and intestine are related”. Front Pharmacol 2022; 13:927384. [PMID: 36160385 PMCID: PMC9489837 DOI: 10.3389/fphar.2022.927384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/22/2022] [Indexed: 11/22/2022] Open
Abstract
Platycodon grandiflorus (Jacq.) A. DC. (PG) root is one of the most commonly used medicine-food materials for respiratory discomfort in Asia, usually in the form of a decoction or leaching solution. As everyone knows, both of decoction and leaching solution is a polyphase dispersion system, containing low-molecular-weight water-soluble active ingredients and hydrophilic macromolecules. This study aimed to discuss the synergistic effect of Platycodon grandiflorus polysaccharide (PGP) and platycodin D (PD) in PG decoction against chronic bronchitis (CB) and the mechanism underlying. A series of PGP, PD, and PGD + PD suspensions were administrated to CB model rats, on the levels of whole animal and in situ intestinal segment with or without mesenteric lymphatic vessels ligation. It exhibited that PGP exhibited synergistic effects with PD, on improving the histopathological abnormity, mucus secretion excess, and immunological imbalance in lung of CB model rat, closely associated with its modulations on the mucosal immunity status in small intestine. The polysaccharide macromolecules in PG decoction or leaching solution should be responsible for the modulation of pulmonary immune state, possibly through the common mucosal immune between small intestine and lung. These results might be a new perspective that illustrates the classical theory of “the lung and intestine are related” in traditional Chinese medicine.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingqing Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Rongrong Ren
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingqing Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Guiming Yan
- School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- *Correspondence: Dengke Yin, ; Ye Yang,
| | - Mingyan Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- School of Nursing, Anhui University of Chinese Medicine, Hefei, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Dengke Yin, ; Ye Yang,
| |
Collapse
|
18
|
Fang H, Wang W, Zhang L, Shen Q, Yuan J, Reichard KK, Hu Z, Medeiros LJ. Reactive Intralymphovascular Immunoblastic Proliferations Mimicking Aggressive Lymphomas. Am J Surg Pathol 2022; 46:326-335. [PMID: 34310368 DOI: 10.1097/pas.0000000000001785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Reactive intralymphovascular immunoblastic proliferations (ILVIPs) may mimic aggressive lymphomas and are rarely reported. Herein, we characterize the clinicopathologic features of 8 patients with ILVIPs. No patients had lymphadenopathy, hepatosplenomegaly, or other findings suggestive of lymphoma. The ILVIPs involved the small or large intestine (n=5) and appendix (n=3). Patients were evaluated for abdominal pain, suspected appendicitis, intestinal obstruction, diverticulitis, volvulus, or tumor resection. Histologic sections showed expanded lymphovascular spaces filled by intermediate to large immunoblasts, positive for CD38, CD43, CD45, CD79a, and MUM1/IRF4 in all cases tested. Five of 6 (83%) cases were positive for CD30. CD20 was weakly positive in a subset of cells in 2 (25%) cases, and PAX5 was weakly positive in 4 (50%) cases. The immunoblasts expressed polytypic light chains in all cases tested. In 1 case, a subset of immunoblasts expressed T-cell markers indicating the presence of a T-cell component. The immunoblasts were negative for ALK, BCL-2, BCL-6, CD10, CD56, CD138, and Epstein-Barr virus-encoded small RNA in all cases assessed. The proliferation index shown by Ki-67 was high with a median of 80%. In all 6 cases tested, the immunoblasts were shown within lymphatic channels highlighted by D2-40. In conclusion, ILVIPs can be rarely observed in patients with inflammatory or infectious conditions, especially in gastrointestinal tract surgical specimens. The immunoblasts are predominantly of B-lineage with a postgerminal center immunophenotype and are located within lymphatic channels. It is essential to distinguish reactive ILVIPs from aggressive lymphomas to avoid unnecessary therapy.
Collapse
Affiliation(s)
- Hong Fang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center
| | - Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Qi Shen
- Department of Pathology, Advent Health-Orlando, Orlando, FL
| | - Ji Yuan
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Kaaren K Reichard
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Zhihong Hu
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
19
|
Zheng R, Huang YM, Zhou Q. Xueshuantong Improves Functions of Lymphatic Ducts and Modulates Inflammatory Responses in Alzheimer's Disease Mice. Front Pharmacol 2021; 12:605814. [PMID: 34650426 PMCID: PMC8505705 DOI: 10.3389/fphar.2021.605814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
Recent studies have revealed significant contributions of lymphatic vessels (LVs) to vital functions of the brain, especially related to clearance of waste from the brain and immune responses in the brain. These studies collectively indicate that enhancing the functions of LVs may improve brain functions during brain aging and in Alzheimer’s disease (AD) where LV functions are impaired. However, it is currently unknown whether this enhancement can be achieved using small molecules. We have previously shown that a widely used Chinese herbal medicine Xueshuantong (XST) significantly improves functions and reduces pathology in AD transgenic mice associated with elevated cerebral blood flow (CBF). Here, we show that XST partially rescues deficits in lymphatic structures, improves clearance of amyloid-β (Aβ) from the brain, and reduces the inflammatory responses in the serum and brains of transgenic AD mice. In addition, we showed that this improvement in the lymphatic system occurs independently of elevated CBF, suggesting independent modulation and limited interaction between blood circulation and lymphatic systems. Moreover, XST treatment leads to a significant increase in GLT-1 level and a significantly lower level of MMP-9 and restores AQP4 polarity in APP/PS1 mice. These results provide the basis for further exploration of XST to enhance or restore LV functions, which may be beneficial to treat neurodegenerative diseases or promote healthy aging.
Collapse
Affiliation(s)
- Rui Zheng
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yang-Mei Huang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
20
|
Creed HA, Rutkowski JM. Emerging roles for lymphatics in acute kidney injury: Beneficial or maleficent? Exp Biol Med (Maywood) 2021; 246:845-850. [PMID: 33467886 DOI: 10.1177/1535370220983235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury, a sudden decline in renal filtration, is a surprisingly common pathology resulting from ischemic events, local or systemic infection, or drug-induced toxicity in the kidney. Unchecked, acute kidney injury can progress to renal failure and even recovered acute kidney injury patients are at an increased risk for developing future chronic kidney disease. The initial extent of inflammation, the specific immune response, and how well inflammation resolves are likely determinants in acute kidney injury-to-chronic kidney disease progression. Lymphatic vessels and their roles in fluid, solute, antigen, and immune cell transport make them likely to have a role in the acute kidney injury response. Lymphatics have proven to be an attractive target in regulating inflammation and immunomodulation in other pathologies: might these strategies be employed in acute kidney injury? Acute kidney injury studies have identified elevated levels of lymphangiogenic ligands following acute kidney injury, with an expansion of the lymphatics in several models post-injury. Manipulating the lymphatics in acute kidney injury, by augmenting or inhibiting their growth or through targeting lymphatic-immune interactions, has met with a range of positive, negative, and sometimes inconclusive results. This minireview briefly summarizes the findings of lymphatic changes and lymphatic roles in the inflammatory response in the kidney following acute kidney injury to discuss whether renal lymphatics are a beneficial, maleficent, or a passive contributor to acute kidney injury recovery.
Collapse
Affiliation(s)
- Heidi A Creed
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| | - Joseph M Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| |
Collapse
|
21
|
Sazhin AV, Nechay TV, Titkova SM, Petukhov VA, Tyagunov AE, Stradymov EA, Ermakov IV, Mishakina NY. [Appendectomy technique: paradigm shift or a well-forgotten old one? The role of mesoappendectomy in prevention of infectious intra-abdominal complications (announcement of RCT)]. Khirurgiia (Mosk) 2020:49-59. [PMID: 33047586 DOI: 10.17116/hirurgia202010149] [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: 11/17/2022]
Abstract
OBJECTIVE To analyze the role of mesoappendixectomy in the development of intra-abdominal surgical site infection (IAB SSI) after LAE. MATERIAL AND METHODS A prospective randomized non-blind multiple-center registered (ClinicalTrials.gov NCT03754777) study has been performed for the period from 2016 to 2018. The study was devoted to effectiveness and safety of the modified enhanced recovery protocol in LAE. In the main group, this protocol (n=56) included routine mesoappendixectomy, restrictive strategy for abdominal drainage and postoperative antibiotic prevention. In the control group (n=71), mesoappendixectomy was performed only in case of necrotic changes. Both groups were comparable by demographic parameters and severity of comorbidities. RESULTS In the main group, significant decrease in the incidence of IAB SSI was found (0% versus 9.8%). Moreover, the main group was characterized by reduced length of hospital-stay (1.43±1.34 d versus 2.94±2, 43 days). CONCLUSION Mesoappendixectomy should be evaluated in further research as a potential factor in prevention of IAB SSI.
Collapse
Affiliation(s)
- A V Sazhin
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - T V Nechay
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - S M Titkova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - V A Petukhov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A E Tyagunov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - E A Stradymov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - I V Ermakov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - N Yu Mishakina
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
22
|
Lai PY, Chang CH, Su HR, Kuo WC. Lymphatic vessel segmentation in optical coherence tomography by adding U-Net-based CNN for artifact minimization. BIOMEDICAL OPTICS EXPRESS 2020; 11:2679-2693. [PMID: 32499952 PMCID: PMC7249833 DOI: 10.1364/boe.389373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/24/2020] [Accepted: 04/16/2020] [Indexed: 06/11/2023]
Abstract
The lymphatic system branches throughout the body to transport bodily fluid and plays a key immune-response role. Optical coherence tomography (OCT) is an emerging technique for the noninvasive and label-free imaging of lymphatic capillaries utilizing low scattering features of the lymph fluid. Here, the proposed lymphatic segmentation method combines U-Net-based CNN, a Hessian vesselness filter, and a modified intensity-thresholding to search the nearby pixels based on the binarized Hessian mask. Compared to previous approaches, the method can extract shapes more precisely, and the segmented result contains minimal artifacts, achieves the dice coefficient of 0.83, precision of 0.859, and recall of 0.803.
Collapse
Affiliation(s)
- Pei-Yu Lai
- Department of Biophotonics, National Yang-Ming University, 155, Sec-2, Li-Nong Street, Taipei 112, Taiwan
| | - Chung-Hsing Chang
- Skin Institute, Hualien Tzu Chi Hospital, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Hong-Ren Su
- Super Genius AItek Co., Ltd, New Taipei City, Taiwan
| | - Wen-Chuan Kuo
- Department of Biophotonics, National Yang-Ming University, 155, Sec-2, Li-Nong Street, Taipei 112, Taiwan
| |
Collapse
|
23
|
Ungaro F, Garlatti V, Massimino L, Spinelli A, Carvello M, Sacchi M, Spanò S, Colasante G, Valassina N, Vetrano S, Malesci A, Peyrin-Biroulet L, Danese S, D'Alessio S. mTOR-Dependent Stimulation of IL20RA Orchestrates Immune Cell Trafficking through Lymphatic Endothelium in Patients with Crohn's Disease. Cells 2019; 8:cells8080924. [PMID: 31426584 PMCID: PMC6721646 DOI: 10.3390/cells8080924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022] Open
Abstract
Crohn’s disease (CD) is a chronic inflammatory condition that can affect different portions of the gastrointestinal tract. Lymphatic drainage was demonstrated to be dysfunctional in CD pathogenesis, ultimately causing the failure of the resolution of intestinal inflammation. To investigate the molecular mechanisms underlying these dysfunctions, we isolated human intestinal lymphatic endothelial cells (HILECs) from surgical specimens of patients undergoing resection for complicated CD (CD HILEC) and from a disease-free margin of surgical specimens of patients undergoing resection for cancer (healthy HILEC). Both cell types underwent transcriptomic profiling, and their barrier functionality was tested using a transwell-based co-culture system between HILEC and lamina propria mononuclear cells (LPMCs). Results showed CD HILEC displayed a peculiar transcriptomic signature that highlighted mTOR signaling as an orchestrator of leukocyte trafficking through the lymphatic barrier of CD patients. Moreover, we demonstrated that LPMC transmigration through the lymphatic endothelium of patients with CD depends on the capability of mTOR to trigger interleukin 20 receptor subunit α (IL20RA)-mediated intracellular signaling. Conclusively, our study suggests that leukocyte trafficking through the intestinal lymphatic microvasculature can be controlled by modulating IL20RA, thus leading to the resolution of chronic inflammation in patients with CD.
Collapse
Affiliation(s)
- Federica Ungaro
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Valentina Garlatti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Luca Massimino
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Michele Carvello
- Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Matteo Sacchi
- Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Salvatore Spanò
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Gaia Colasante
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Nicholas Valassina
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefania Vetrano
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Alberto Malesci
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 200129 Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Laurent Peyrin-Biroulet
- Inserm Ngere and Nancy University Hospital, Lorraine University, 54500 Vandoeuvre-lès-Nancy, France
| | - Silvio Danese
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Silvia D'Alessio
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy.
| |
Collapse
|
24
|
Morfoisse F, Noel A. Lymphatic and blood systems: Identical or fraternal twins? Int J Biochem Cell Biol 2019; 114:105562. [PMID: 31278994 DOI: 10.1016/j.biocel.2019.105562] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
Blood and lymphatic systems work in close collaboration to ensure their respective physiological functions. The lymphatic vessel network is being extensively studied, but has been overlooked as compared to the blood vasculature mainly due to the problematic discrimination of lymphatic vessels from the blood ones. This issue has been fortunately resolved in the past decade leading to the emergence of a huge amount of data in lymphatic biology revealing many shared features with the blood vasculature. However, this likeliness between the two vascular systems may lead to a simplistic view of lymphatics and a direct transcription of what is known for the blood system to the lymphatic one, thereby neglecting the lymphatic specificities. In this context, this review aims to clarify the main differences between the two vascular systems focusing on recently discovered lymphatic features.
Collapse
Affiliation(s)
- Florent Morfoisse
- Laboratory of Tumor and Development Biology, GIGA (GIGA-Cancer), Liege University, B23, Avenue Hippocrate 13, 4000, Liege, Belgium
| | - Agnès Noel
- Laboratory of Tumor and Development Biology, GIGA (GIGA-Cancer), Liege University, B23, Avenue Hippocrate 13, 4000, Liege, Belgium.
| |
Collapse
|
25
|
Baseline Lymphatic Dysfunction Amplifies the Negative Effects of Lymphatic Injury. Plast Reconstr Surg 2019; 143:77e-87e. [PMID: 30589786 DOI: 10.1097/prs.0000000000005091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Genetic mutations and obesity increase the risk of secondary lymphedema, suggesting that impaired lymphatic function before surgical injury may contribute to disease pathophysiology. Previous studies show that obesity not only decreases lymphatic function, but also markedly increases pathologic changes, such as swelling, fibroadipose deposition, and inflammation. However, although these reports provide circumstantial evidence supporting the hypothesis that baseline lymphatic defects amplify the effect of lymphatic injury, the mechanisms regulating this association remain unknown. METHODS Baseline lymphatic morphology, leakiness, pumping, immune cell trafficking, and local inflammation and fibroadipose deposition were assessed in wild-type and Prox1-haploinsufficient (Prox1) mice, which have previously been shown to have abnormal vasculature without overt evidence of lymphedema. In subsequent experiments, wild-type and Prox1 mice underwent popliteal lymph node dissection to evaluate the effect of lymphatic injury. Repeated testing of all variables was conducted 4 weeks postoperatively. RESULTS At baseline, Prox1 mice had dilated, leaky lymphatic vessels corresponding to low-grade inflammation and decreased pumping and transport function, compared with wild-type mice. Popliteal lymph node dissection resulted in evidence of lymphedema in both Prox1 and wild-type mice, but popliteal lymph node dissection-treated Prox1 mice had increased inflammation and decreased lymphatic pumping. CONCLUSIONS Subclinical lymphatic dysfunction exacerbates the pathologic changes of lymphatic injury, an effect that is multifactorial and related to increased lymphatic leakiness, perilymphatic accumulation of inflammatory cells, and impaired pumping and transport capacity. These findings suggest that preoperative testing of lymphatic function may enable clinicians to more accurately risk-stratify patients and design targeted preventative strategies.
Collapse
|
26
|
Witzel II, Nasser R, Garcia-Sabaté A, Sapudom J, Ma C, Chen W, Teo JCM. Deconstructing Immune Microenvironments of Lymphoid Tissues for Reverse Engineering. Adv Healthc Mater 2019; 8:e1801126. [PMID: 30516005 DOI: 10.1002/adhm.201801126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Indexed: 01/01/2023]
Abstract
The immune microenvironment presents a diverse panel of cues that impacts immune cell migration, organization, differentiation, and the immune response. Uniquely, both the liquid and solid phases of every specific immune niche within the body play an important role in defining cellular functions in immunity at that particular location. The in vivo immune microenvironment consists of biomechanical and biochemical signals including their gradients, surface topography, dimensionality, modes of ligand presentation, and cell-cell interactions, and the ability to recreate these immune biointerfaces in vitro can provide valuable insights into the immune system. This manuscript reviews the critical roles played by different immune cells and surveys the current progress of model systems for reverse engineering of immune microenvironments with a focus on lymphoid tissues.
Collapse
Affiliation(s)
- Ini-Isabée Witzel
- Core Technology Platforms; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Rasha Nasser
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
- Department of Biomedical Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| |
Collapse
|
27
|
Vuorio T, Ylä-Herttuala E, Laakkonen JP, Laidinen S, Liimatainen T, Ylä-Herttuala S. Downregulation of VEGFR3 signaling alters cardiac lymphatic vessel organization and leads to a higher mortality after acute myocardial infarction. Sci Rep 2018; 8:16709. [PMID: 30420641 PMCID: PMC6232169 DOI: 10.1038/s41598-018-34770-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 10/26/2018] [Indexed: 02/02/2023] Open
Abstract
Heart has a wide lymphatic network but the importance of cardiac lymphatic system in heart diseases has remained unclear. Vascular Endothelial Growth Factor Receptor 3 (VEGFR3) is a key molecule in the development and maintenance of cardiac lymphatic vessels. Here we characterized the role of VEGFR3 in healthy hearts and after myocardial infarction (MI) by using sVEGFR3 transgenic mice expressing a soluble decoy VEGFR3 under K14 promoter and Chy mice which have an inactivating mutation in the VEGFR3 gene. Cardiac lymphatic vessels were significantly dilated in the healthy hearts of sVEGFR3 mice when compared to controls. Lymphatic vessels formed large sheet-like structures in Chy mice. Attenuated VEGFR3 signaling led to a more severe MI predisposing to a significantly higher mortality in sVEGFR3 mice than in control mice. sVEGFR3 mice displayed intramyocardial hemorrhages in the infarcted area indicating hyperpermeability of the vasculature. Furthermore, novel MRI methods TRAFF2 and TRAFF4 and histological analysis revealed a modified structure of the fibrotic infarcted area in sVEGFR3 mice. In conclusion, the downregulation of VEGFR3 signaling modifies the structure of cardiac lymphatic network and causes vascular leakiness and increased mortality after MI.
Collapse
Affiliation(s)
- Taina Vuorio
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Elias Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Svetlana Laidinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Timo Liimatainen
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland.,Department of Diagnostic Radiology, University Hospital of Oulu, P.O. Box 50, FI-90029 OYS, Oulu, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland. .,Heart Center and Gene Therapy Unit, Kuopio University Hospital, P.O. Box 1777, FI-70211, Kuopio, Finland.
| |
Collapse
|
28
|
|
29
|
Jung YJ, Park Y, Kim HS, Lee HJ, Kim YN, Lee J, Kim YH, Maeng YS, Kwon JY. Abnormal lymphatic vessel development is associated with decreased decidual regulatory T cells in severe preeclampsia. Am J Reprod Immunol 2018; 80:e12970. [PMID: 29756666 DOI: 10.1111/aji.12970] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/09/2018] [Indexed: 12/29/2022] Open
Abstract
PROBLEM The lymphatic vasculature controls leukocytes trafficking and limits the adaptive immune response. In previous models of preeclampsia (PE), defective immune function caused by disruption of lymphangiogenesis was shown to be involved in the disease pathophysiology. Especially, the dysfunction of regulatory T cells (Treg) at the maternal-fetal interface may be one of the causes of severe PE. In particular, activation of Tregs to obtain immune tolerance requires adequate antigen presentation through the lymphatic system. We hypothesized that impaired lymphangiogenesis and imbalanced Tregs at the maternal-fetal interface are associated with the pathophysiology of severe PE. However, the current research addressing this hypothesis is limited. Therefore, to compare differences in lymphangiogenesis in severe PE and normal conditions, we aimed to examine the location of lymphatics at the maternal-fetal interface and to investigate the association between lymphangiogenesis and Tregs in severe PE. METHOD OF STUDY We obtained entire uterus from normal pregnant mice. Placental and fetal membranes, including decidua, were obtained from 10 pregnant women with severe PE and 10 gestational age-matched controls. Immunohistochemistry for LYVE1 was used to localize the distribution of lymphatic vessels and CD4, CD25, and FOXP3 for Treg. RESULTS LYVE1-positive vessels were present in the uterine wall of mice. LYVE1-positive lymphatic vessels were localized on the human decidua. Tubular lymphatics were abundant in the control decidua, but significantly reduced in severe PE. Furthermore, lymphatic vessel density correlated with the number of decidual Tregs. CONCLUSION Abnormal decidual lymphangiogenesis is associated with reduced numbers of decidual Tregs in severe PE.
Collapse
Affiliation(s)
- Yun Ji Jung
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Yejin Park
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Hyun-Soo Kim
- Department of Pathology, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Hwa Jin Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Yoo-Na Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - JoonHo Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Young-Han Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - Ja-Young Kwon
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| |
Collapse
|
30
|
Ge Y, Li Y, Gong J, Zhu W. Mesenteric organ lymphatics and inflammatory bowel disease. Ann Anat 2018; 218:199-204. [PMID: 29723582 DOI: 10.1016/j.aanat.2018.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/14/2018] [Accepted: 03/01/2018] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel disease (IBD) is a complex gastrointestinal disorder and its etiology is unclear yet. Current theory in IBD is focused on genetics, immunity and intestinal microbes. Emerging clinical evidence and experimental results suggest that morphologic abnormalities and dysfunction of mesenteric lymphatics may have potential roles in the pathogenesis and disease course of IBD. In this review, we summarize the findings of specific investigations of the lymphatics and explore its role in IBD.
Collapse
Affiliation(s)
- Yuanyuan Ge
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002 PR China
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002 PR China.
| | - Jianfeng Gong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002 PR China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002 PR China
| |
Collapse
|
31
|
Kou CTJ, Kandpal RP. Differential Expression Patterns of Eph Receptors and Ephrin Ligands in Human Cancers. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7390104. [PMID: 29682554 PMCID: PMC5851329 DOI: 10.1155/2018/7390104] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/11/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
Eph receptors constitute the largest family of receptor tyrosine kinases, which are activated by ephrin ligands that either are anchored to the membrane or contain a transmembrane domain. These molecules play important roles in the development of multicellular organisms, and the physiological functions of these receptor-ligand pairs have been extensively documented in axon guidance, neuronal development, vascular patterning, and inflammation during tissue injury. The recognition that aberrant regulation and expression of these molecules lead to alterations in proliferative, migratory, and invasive potential of a variety of human cancers has made them potential targets for cancer therapeutics. We present here the involvement of Eph receptors and ephrin ligands in lung carcinoma, breast carcinoma, prostate carcinoma, colorectal carcinoma, glioblastoma, and medulloblastoma. The aberrations in their abundances are described in the context of multiple signaling pathways, and differential expression is suggested as the mechanism underlying tumorigenesis.
Collapse
Affiliation(s)
- Chung-Ting Jimmy Kou
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Raj P. Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
32
|
Lerner TR, Queval CJ, Fearns A, Repnik U, Griffiths G, Gutierrez MG. Phthiocerol dimycocerosates promote access to the cytosol and intracellular burden of Mycobacterium tuberculosis in lymphatic endothelial cells. BMC Biol 2018; 16:1. [PMID: 29325545 PMCID: PMC5795283 DOI: 10.1186/s12915-017-0471-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023] Open
Abstract
Background Phthiocerol dimycocerosates (PDIM), glycolipids found on the outer surface of virulent members of the Mycobacterium tuberculosis (Mtb) complex, are a major contributing factor to the pathogenesis of Mtb. Myelocytic cells, such as macrophages and dendritic cells, are the primary hosts for Mtb after infection and previous studies have shown multiple roles for PDIM in supporting Mtb in these cells. However, Mtb can infect other cell types. We previously showed that Mtb efficiently replicates in human lymphatic endothelial cells (hLECs) and that the hLEC cytosol acts as a reservoir for Mtb in humans. Here, we examined the role of PDIM in Mtb translocation to the cytosol in hLECs. Results Analysis of a Mtb mutant unable to produce PDIM showed less co-localisation of bacteria with the membrane damage marker Galectin-8 (Gal8), indicating that PDIM strongly contribute to phagosomal membrane damage. Lack of this Mtb lipid also leads to a reduction in the proportion of Mtb co-localising with markers of macroautophagic removal of intracellular bacteria (xenophagy) such as ubiquitin, p62 and NDP52. hLEC imaging with transmission electron microscopy shows that Mtb mutants lacking PDIM are much less frequently localised in the cytosol, leading to a lower intracellular burden. Conclusions PDIM is needed for the disruption of the phagosome membrane in hLEC, helping Mtb avoid the hydrolytic phagolysosomal milieu. It facilitates the translocation of Mtb into the cytosol, and the decreased intracellular burden of Mtb lacking PDIM indicates that the cytosol is the preferred replicative niche for Mtb in these cells. We hypothesise that pharmacological targeting of PDIM synthesis in Mtb would reduce the formation of a lymphatic reservoir of Mtb in humans.
Collapse
Affiliation(s)
- Thomas R Lerner
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | | | - Antony Fearns
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371, Oslo, Norway
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371, Oslo, Norway
| | | |
Collapse
|
33
|
Blei F. Update June 2017. Lymphat Res Biol 2017. [DOI: 10.1089/lrb.2017.29024.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|