1
|
Karakousi T, Cristaldi V, Lopes de Oliveira ML, Medeiros Geraldo LH, González-Robles TJ, da Silva G, Breazeale AP, Encarnacion-Rosado J, Pozniak J, Kimmelman AC, Ruggles KV, Chris Marine J, Chandel NS, Lund AW. IFNγ-dependent metabolic reprogramming restrains an immature, pro-metastatic lymphatic state in melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626426. [PMID: 39677662 PMCID: PMC11642899 DOI: 10.1101/2024.12.02.626426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Lymphatic vessels play a crucial role in activating anti-tumor immune surveillance but also contribute to metastasis and systemic tumor progression. Whether distinct lymphatic phenotypes exist that govern the switch between immunity and metastasis remains unclear. Here we reveal that cytotoxic immunity normalizes lymphatic function and uncouples immune and metastatic potential. We find that in mice and humans, intratumoral lymphatic vessel density negatively correlates with productive cytotoxic immune responses and identify IFNγ as an intrinsic inhibitor of lymphangiogenesis. Specific deletion of the Ifngr1 in lymphatic endothelial cells (LECs) greatly expanded the intratumoral lymphatic network and drove the emergence of a tip-like endothelial state, promoting lymph node metastasis but not dendritic cell migration. IFNγ inhibits oxidative phosphorylation, which is required for proliferation and acquisition of the pathologic transcriptional state. Our data indicate that IFNγ induces a phenotypic switch in tumor-associated lymphatic vessels to reinforce canonical immune surveillance and block metastasis.
Collapse
Affiliation(s)
- Triantafyllia Karakousi
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vanessa Cristaldi
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Luiz Henrique Medeiros Geraldo
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY
| | - Tania J. González-Robles
- Institute of Systems Genetics, Department of Precision Medicine, NYU Grossman School of Medicine, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY USA
| | - Gabrielle da Silva
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Alec P. Breazeale
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Joel Encarnacion-Rosado
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, NY, USA
| | - Joanna Pozniak
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alec C. Kimmelman
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Kelly V. Ruggles
- Institute of Systems Genetics, Department of Precision Medicine, NYU Grossman School of Medicine, New York, NY USA
| | - J. Chris Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Amanda W. Lund
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| |
Collapse
|
2
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza induces lung lymphangiogenesis independent of YAP/TAZ activity in lymphatic endothelial cells. Sci Rep 2024; 14:21324. [PMID: 39266641 PMCID: PMC11393066 DOI: 10.1038/s41598-024-72115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA.
| | - Senegal Carty
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Fengzhi Shao
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Jhonatan Henao-Vasquez
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alexandra B Ysasi
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Michelle Zeng
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Anne Hinds
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Ming Lo
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Comparative Pathology Laboratory, Boston University National Emerging and Infectious Disease Laboratories, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alan Fine
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| |
Collapse
|
3
|
Wei R, Zheng Z, Li Q, Qian Y, Wu C, Li Y, Wang M, Chen J, He W. Prognostic and predictive value of examined lymph node count in stage III colorectal cancer: a population based study. World J Surg Oncol 2024; 22:155. [PMID: 38872183 PMCID: PMC11170906 DOI: 10.1186/s12957-024-03404-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND The role of tumor-draining lymph nodes in the progression of malignant tumors, including stage III colorectal cancer (CRC), is critical. However, the prognostic and predictive value of the number of examined lymph nodes (ELNs) are not fully understood. METHODS This population-based study retrospectively analyzed data from 106,843 patients with stage III CRC who underwent surgical treatment and registered in three databases from 2004 to 2021. The Surveillance, Epidemiology, and End Results (SEER) cohort was divided using into training and test cohorts at a ratio of 3:2. We employed restricted cubic spline (RCS) curves to explore nonlinear relationships between overall survival (OS) and ELNs counts and performed Cox regression to evaluate hazard ratios across different ELNs count subtypes. Additional validation cohorts were utilized from the First Affiliated Hospital, Sun Yat-sen University and The Cancer Genome Atlas (TCGA) under the same criteria. Outcomes measured included OS, cancer-specific survival (CSS), and progression-free survival (PFS). Molecular analyses involved differential gene expression using the "limma" package and immune profiling through CIBERSORT. Tissue microarray slides and multiplex immunofluorescence (MIF) were used to assess protein expression and immune cell infiltration. RESULTS Patients with higher ELNs counts (≥ 17) demonstrated significantly better long-term survival outcomes across all cohorts. Enhanced OS, CSS, and PFS were notably evident in the LN-ELN group compared to those with fewer ELNs. Cox regression models underscored the prognostic value of higher ELNs counts across different patient subgroups by age, sex, tumor differentiation, and TNM stages. Subtype analysis based on ELNs count revealed a marked survival benefit in patients treated with adjuvant chemotherapy in the medium and large ELNs counts (≥ 12), whereas those with fewer ELNs showed negligible benefits. RNA sequencing and MIF indicated elevated immune activation in the LN-ELN group, characterized by increased CD3+, CD4+, and CD8 + T cells within the tumor microenvironment. CONCLUSIONS The number of ELNs independently predicts survival and the immunological landscape at the tumor site in stage III CRC, underscoring its dual prognostic and predictive value.
Collapse
Affiliation(s)
- Ran Wei
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zifan Zheng
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qinghai Li
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan Qian
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Chong Wu
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yin Li
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Mian Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Jianhui Chen
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, China.
| | - Weiling He
- Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
4
|
Heim TA, Schultz AC, Delclaux I, Cristaldi V, Churchill MJ, Ventre KS, Lund AW. Lymphatic vessel transit seeds cytotoxic resident memory T cells in skin draining lymph nodes. Sci Immunol 2024; 9:eadk8141. [PMID: 38848340 DOI: 10.1126/sciimmunol.adk8141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/14/2024] [Indexed: 06/09/2024]
Abstract
Lymphatic transport shapes the homeostatic immune repertoire of lymph nodes (LNs). LN-resident memory T cells (TRMs) play an important role in site-specific immune memory, yet how LN TRMs form de novo after viral infection remains unclear. Here, we tracked the anatomical distribution of antiviral CD8+ T cells as they seeded skin and LN TRMs using a model of vaccinia virus-induced skin infection. LN TRMs localized to the draining LNs (dLNs) of infected skin, and their formation depended on the lymphatic egress of effector CD8+ T cells from the skin, already poised for residence. Effector CD8+ T cell transit through skin was required to populate LN TRMs in dLNs, a process reinforced by antigen encounter in skin. Furthermore, LN TRMs were protective against viral rechallenge in the absence of circulating memory T cells. These data suggest that a subset of tissue-infiltrating CD8+ T cells egress from tissues during viral clearance and establish a layer of regional protection in the dLN basin.
Collapse
Affiliation(s)
- Taylor A Heim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Austin C Schultz
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ines Delclaux
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vanessa Cristaldi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Madeline J Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Katherine S Ventre
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
5
|
Karakousi T, Mudianto T, Lund AW. Lymphatic vessels in the age of cancer immunotherapy. Nat Rev Cancer 2024; 24:363-381. [PMID: 38605228 DOI: 10.1038/s41568-024-00681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/13/2024]
Abstract
Lymphatic transport maintains homeostatic health and is necessary for immune surveillance, and yet lymphatic growth is often associated with solid tumour development and dissemination. Although tumour-associated lymphatic remodelling and growth were initially presumed to simply expand a passive route for regional metastasis, emerging research puts lymphatic vessels and their active transport at the interface of metastasis, tumour-associated inflammation and systemic immune surveillance. Here, we discuss active mechanisms through which lymphatic vessels shape their transport function to influence peripheral tissue immunity and the current understanding of how tumour-associated lymphatic vessels may both augment and disrupt antitumour immune surveillance. We end by looking forward to emerging areas of interest in the field of cancer immunotherapy in which lymphatic vessels and their transport function are likely key players: the formation of tertiary lymphoid structures, immune surveillance in the central nervous system, the microbiome, obesity and ageing. The lessons learnt support a working framework that defines the lymphatic system as a key determinant of both local and systemic inflammatory networks and thereby a crucial player in the response to cancer immunotherapy.
Collapse
Affiliation(s)
- Triantafyllia Karakousi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
6
|
Hsu J, Kim S, Anandasabapathy N. Vaccinia Virus: Mechanisms Supporting Immune Evasion and Successful Long-Term Protective Immunity. Viruses 2024; 16:870. [PMID: 38932162 PMCID: PMC11209207 DOI: 10.3390/v16060870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Vaccinia virus is the most successful vaccine in human history and functions as a protective vaccine against smallpox and monkeypox, highlighting the importance of ongoing research into vaccinia due to its genetic similarity to other emergent poxviruses. Moreover, vaccinia's ability to accommodate large genetic insertions makes it promising for vaccine development and potential therapeutic applications, such as oncolytic agents. Thus, understanding how superior immunity is generated by vaccinia is crucial for designing other effective and safe vaccine strategies. During vaccinia inoculation by scarification, the skin serves as a primary site for the virus-host interaction, with various cell types playing distinct roles. During this process, hematopoietic cells undergo abortive infections, while non-hematopoietic cells support the full viral life cycle. This differential permissiveness to viral replication influences subsequent innate and adaptive immune responses. Dendritic cells (DCs), key immune sentinels in peripheral tissues such as skin, are pivotal in generating T cell memory during vaccinia immunization. DCs residing in the skin capture viral antigens and migrate to the draining lymph nodes (dLN), where they undergo maturation and present processed antigens to T cells. Notably, CD8+ T cells are particularly significant in viral clearance and the establishment of long-term protective immunity. Here, we will discuss vaccinia virus, its continued relevance to public health, and viral strategies permissive to immune escape. We will also discuss key events and populations leading to long-term protective immunity and remaining key gaps.
Collapse
Affiliation(s)
- Joy Hsu
- Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Department of Dermatology, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Suyon Kim
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Niroshana Anandasabapathy
- Department of Dermatology, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
7
|
Mao X, Tang X, Pan H, Yu M, Ji S, Qiu W, Che N, Zhang K, Huang Z, Jiang Y, Wang J, Zhong Z, Wang J, Liu M, Chen M, Zhou W, Wang S. B Cells and IL-21-Producing Follicular Helper T Cells Cooperate to Determine the Dynamic Alterations of Premetastatic Tumor Draining Lymph Nodes of Breast Cancer. RESEARCH (WASHINGTON, D.C.) 2024; 7:0346. [PMID: 38559676 PMCID: PMC10981934 DOI: 10.34133/research.0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Metastasis is the major cause of cancer-related death, and lymph node is the most common site of metastasis in breast cancer. However, the alterations that happen in tumor-draining lymph nodes (TDLNs) to form a premetastatic microenvironment are largely unknown. Here, we first report the dynamic changes in size and immune status of TDLNs before metastasis in breast cancer. With the progression of tumor, the TDLN is first enlarged and immune-activated at early stage that contains specific antitumor immunity against metastasis. The TDLN is then contracted and immunosuppressed at late stage before finally getting metastasized. Mechanistically, B and follicular helper T (Tfh) cells parallelly expand and contract to determine the size of TDLN. The activation status and specific antitumor immunity of CD8+ T cells in the TDLN are determined by interleukin-21 (IL-21) produced by Tfh cells, thus showing parallel changes. The turn from activated enlargement to suppressed contraction is due to the spontaneous contraction of germinal centers mediated by follicular regulatory T cells. On the basis of the B-Tfh-IL-21-CD8+ T cell axis, we prove that targeting the axis could activate TDLNs to resist metastasis. Together, our findings identify the dynamic alterations and regulatory mechanisms of premetastatic TDLNs of breast cancer and provide new strategies to inhibit lymph node metastasis.
Collapse
Affiliation(s)
- Xinrui Mao
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Xinyu Tang
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Hong Pan
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Muxin Yu
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Sihan Ji
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Wen Qiu
- Department of Immunology,
Nanjing Medical University, Nanjing 211166, China
| | - Nan Che
- Department of Rheumatology and Immunology,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Kai Zhang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
- Pancreatic Center & Department of General Surgery,
The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, Jiangsu, China
- Pancreas Institute of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhendong Huang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
- Department of Pathology,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yunshan Jiang
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Ji Wang
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Zhaoyun Zhong
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Jiaming Wang
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Mingduo Liu
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Mingkang Chen
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
- Department of Ophthalmology,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wenbin Zhou
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| | - Shui Wang
- Department of Breast Surgery,
The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health,
Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
8
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza Induces Lung Lymphangiogenesis Independent of YAP/TAZ Activity in Lymphatic Endothelial Cells. RESEARCH SQUARE 2024:rs.3.rs-3951689. [PMID: 38463972 PMCID: PMC10925403 DOI: 10.21203/rs.3.rs-3951689/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases 2-fold at 7 days post-influenza infection (dpi) and 3-fold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Boston University Chobanian and Avedisian School of Medicine
| | - Senegal Carty
- Boston University Chobanian and Avedisian School of Medicine
| | - Fengzhi Shao
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Michelle Zeng
- Boston University Chobanian and Avedisian School of Medicine
| | - Anne Hinds
- Boston University Chobanian and Avedisian School of Medicine
| | - Ming Lo
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Matthew R Jones
- Boston University Chobanian and Avedisian School of Medicine
| | - Alan Fine
- Boston University Chobanian and Avedisian School of Medicine
| |
Collapse
|
9
|
Gong H, Wang T, Sun X, Zhang Y, Qiu Y, Sun W, Zhang Y, Teng P, Hu Y, Hu X, Ma L, Xu Q, Zhao H. Fibroblasts facilitate lymphatic vessel formation in transplanted heart. Theranostics 2024; 14:1886-1908. [PMID: 38505621 PMCID: PMC10945330 DOI: 10.7150/thno.92103] [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: 11/10/2023] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
Rationale: Lymphangiogenesis plays a critical role in the transplanted heart. The remodeling of lymphatics in the transplanted heart and the source of newly formed lymphatic vessels are still controversial, especially the mechanism of lymphangiogenesis remains limited. Methods: Heart transplantation was performed among BALB/c, C57BL/6J, Cag-Cre, Lyve1-CreERT2;Rosa26-tdTomato and Postn(2A-CreERT2-wpre-pA)1;Rosa26-DTA mice. scRNA-seq, Elisa assay, Western blotting, Q-PCR and immunohistochemical staining were used to identify the cells and cell-cell communications of allograft heart. Cell depletion was applied to in vivo and in vitro experiments. Whole-mount staining and three-dimensional reconstruction depicted the cell distribution within transparent transplanted heart. Results: Genetic lineage tracing mice and scRNA-seq analysis have revealed that these newly formed lymphatic vessels mainly originate from recipient LYVE1+ cells. It was found that LECs primarily interact with activated fibroblasts. Inhibition of lymphatic vessel formation using a VEGFR3 inhibitor resulted in a decreased survival time of transplanted hearts. Furthermore, when activated fibroblasts were ablated in transplanted hearts, there was a significant suppression of lymphatic vessel generation, leading to earlier graft failure. Additional investigations have shown that activated fibroblasts promote tube formation of LECs primarily through the activation of various signaling pathways, including VEGFD/VEGFR3, MDK/NCL, and SEMA3C/NRP2. Interestingly, knockdown of VEGFD and MDK in activated fibroblasts impaired cardiac lymphangiogenesis after heart transplantation. Conclusions: Our study indicates that cardiac lymphangiogenesis primarily originates from recipient cells, and activated fibroblasts play a crucial role in facilitating the generation of lymphatic vessels after heart transplantation. These findings provide valuable insights into potential therapeutic targets for enhancing graft survival.
Collapse
Affiliation(s)
- Hui Gong
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Wang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaotong Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuesheng Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yichao Qiu
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenlong Sun
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
| | - Yue Zhang
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
| | - Peng Teng
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Ma
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haige Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Thim EA, Kitelinger LE, Rivera-Escalera F, Mathew AS, Elliott MR, Bullock TNJ, Price RJ. Focused ultrasound ablation of melanoma with boiling histotripsy yields abscopal tumor control and antigen-dependent dendritic cell activation. Theranostics 2024; 14:1647-1661. [PMID: 38389838 PMCID: PMC10879863 DOI: 10.7150/thno.92089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/04/2024] [Indexed: 02/24/2024] Open
Abstract
Background: Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods: BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results: BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion: When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Eric A. Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Lydia E. Kitelinger
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Fátima Rivera-Escalera
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Alexander S. Mathew
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael R. Elliott
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
11
|
Femel J, Hill C, Illa Bochaca I, Booth JL, Asnaashari TG, Steele MM, Moshiri AS, Do H, Zhong J, Osman I, Leachman SA, Tsujikawa T, White KP, Chang YH, Lund AW. Quantitative multiplex immunohistochemistry reveals inter-patient lymphovascular and immune heterogeneity in primary cutaneous melanoma. Front Immunol 2024; 15:1328602. [PMID: 38361951 PMCID: PMC10867179 DOI: 10.3389/fimmu.2024.1328602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction Quantitative, multiplexed imaging is revealing complex spatial relationships between phenotypically diverse tumor infiltrating leukocyte populations and their prognostic implications. The underlying mechanisms and tissue structures that determine leukocyte distribution within and around tumor nests, however, remain poorly understood. While presumed players in metastatic dissemination, new preclinical data demonstrates that blood and lymphatic vessels (lymphovasculature) also dictate leukocyte trafficking within tumor microenvironments and thereby impact anti-tumor immunity. Here we interrogate these relationships in primary human cutaneous melanoma. Methods We established a quantitative, multiplexed imaging platform to simultaneously detect immune infiltrates and tumor-associated vessels in formalin-fixed paraffin embedded patient samples. We performed a discovery, retrospective analysis of 28 treatment-naïve, primary cutaneous melanomas. Results Here we find that the lymphvasculature and immune infiltrate is heterogenous across patients in treatment naïve, primary melanoma. We categorized five lymphovascular subtypes that differ by functionality and morphology and mapped their localization in and around primary tumors. Interestingly, the localization of specific vessel subtypes, but not overall vessel density, significantly associated with the presence of lymphoid aggregates, regional progression, and intratumoral T cell infiltrates. Discussion We describe a quantitative platform to enable simultaneous lymphovascular and immune infiltrate analysis and map their spatial relationships in primary melanoma. Our data indicate that tumor-associated vessels exist in different states and that their localization may determine potential for metastasis or immune infiltration. This platform will support future efforts to map tumor-associated lymphovascular evolution across stage, assess its prognostic value, and stratify patients for adjuvant therapy.
Collapse
Affiliation(s)
- Julia Femel
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, United States
| | - Cameron Hill
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Irineu Illa Bochaca
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Jamie L. Booth
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, United States
| | - Tina G. Asnaashari
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health & Science University, Portland, OR, United States
| | - Maria M. Steele
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Ata S. Moshiri
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Hyungrok Do
- Department of Population Health, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Judy Zhong
- Department of Population Health, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| | - Iman Osman
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| | - Sancy A. Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Takahiro Tsujikawa
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, United States
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kevin P. White
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
| | - Young H. Chang
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health & Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Amanda W. Lund
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, United States
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health & Science University, Portland, OR, United States
- Laura and Isaac Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Pathology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
12
|
Thim EA, Kitelinger LE, Rivera-Escalera F, Mathew AS, Elliott MR, Bullock TNJ, Price RJ. Focused ultrasound ablation of melanoma with boiling histotripsy yields abscopal tumor control and antigen-dependent dendritic cell activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.02.552844. [PMID: 37732205 PMCID: PMC10508728 DOI: 10.1101/2023.09.02.552844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Background Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Eric A. Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | | | - Fátima Rivera-Escalera
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | - Alexander S. Mathew
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Michael R. Elliott
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | | | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
13
|
Delclaux I, Ventre KS, Jones D, Lund AW. The tumor-draining lymph node as a reservoir for systemic immune surveillance. Trends Cancer 2024; 10:28-37. [PMID: 37863720 PMCID: PMC10843049 DOI: 10.1016/j.trecan.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023]
Abstract
Early in solid tumor development, antigens are presented in tumor-draining lymph nodes (tdLNs), a process that is necessary to set up immune surveillance. Recent evidence indicates that tdLNs fuel systemic tumor-specific T cell responses which may halt cancer progression and facilitate future responses to immunotherapy. These protective responses, however, are subject to progressive dysfunction exacerbated by lymph node (LN) metastasis. We discuss emerging preclinical and clinical literature indicating that the tdLN is a crucial reservoir for systemic immunity that can potentiate immune surveillance. We also discuss the impact of LN metastasis and argue that a better understanding of the relationship between LN metastasis and systemic immunity will be necessary to direct regional disease management in the era of immunotherapy.
Collapse
Affiliation(s)
- Ines Delclaux
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Katherine S Ventre
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Dennis Jones
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
14
|
Heim TA, Schultz AC, Delclaux I, Cristaldi V, Churchill MJ, Lund AW. Lymphatic vessel transit seeds precursors to cytotoxic resident memory T cells in skin draining lymph nodes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555369. [PMID: 37693469 PMCID: PMC10491166 DOI: 10.1101/2023.08.29.555369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Resident memory T cells (TRM) provide rapid, localized protection in peripheral tissues to pathogens and cancer. While TRM are also found in lymph nodes (LN), how they develop during primary infection and their functional significance remains largely unknown. Here, we track the anatomical distribution of anti-viral CD8+ T cells as they simultaneously seed skin and LN TRM using a model of skin infection with restricted antigen distribution. We find exquisite localization of LN TRM to the draining LN of infected skin. LN TRM formation depends on lymphatic transport and specifically egress of effector CD8+ T cells that appear poised for residence as early as 12 days post infection. Effector CD8+ T cell transit through skin is necessary and sufficient to populate LN TRM in draining LNs, a process reinforced by antigen encounter in skin. Importantly, we demonstrate that LN TRM are sufficient to provide protection against pathogenic rechallenge. These data support a model whereby a subset of tissue infiltrating CD8+ T cells egress during viral clearance, and establish regional protection in the draining lymphatic basin as a mechanism to prevent pathogen spread.
Collapse
Affiliation(s)
- Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Austin C. Schultz
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ines Delclaux
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vanessa Cristaldi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - Amanda W. Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
15
|
Maisel K, McClain CA, Bogseth A, Thomas SN. Nanotechnologies for Physiology-Informed Drug Delivery to the Lymphatic System. Annu Rev Biomed Eng 2023; 25:233-256. [PMID: 37000965 PMCID: PMC10879987 DOI: 10.1146/annurev-bioeng-092222-034906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
Accompanying the increasing translational impact of immunotherapeutic strategies to treat and prevent disease has been a broadening interest across both bioscience and bioengineering in the lymphatic system. Herein, the lymphatic system physiology, ranging from its tissue structures to immune functions and effects, is described. Design principles and engineering approaches to analyze and manipulate this tissue system in nanoparticle-based drug delivery applications are also elaborated.
Collapse
Affiliation(s)
- Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Claire A McClain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA;
| | - Amanda Bogseth
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Steele MM, Jaiswal A, Delclaux I, Dryg ID, Murugan D, Femel J, Son S, du Bois H, Hill C, Leachman SA, Chang YH, Coussens LM, Anandasabapathy N, Lund AW. T cell egress via lymphatic vessels is tuned by antigen encounter and limits tumor control. Nat Immunol 2023; 24:664-675. [PMID: 36849745 PMCID: PMC10998279 DOI: 10.1038/s41590-023-01443-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Antigen-specific CD8+ T cell accumulation in tumors is a prerequisite for effective immunotherapy, and yet the mechanisms of lymphocyte transit are not well defined. Here we show that tumor-associated lymphatic vessels control T cell exit from tumors via the chemokine CXCL12, and intratumoral antigen encounter tunes CXCR4 expression by effector CD8+ T cells. Only high-affinity antigen downregulates CXCR4 and upregulates the CXCL12 decoy receptor, ACKR3, thereby reducing CXCL12 sensitivity and promoting T cell retention. A diverse repertoire of functional tumor-specific CD8+ T cells, therefore, exit the tumor, which limits the pool of CD8+ T cells available to exert tumor control. CXCR4 inhibition or loss of lymphatic-specific CXCL12 boosts T cell retention and enhances tumor control. These data indicate that strategies to limit T cell egress might be an approach to boost the quantity and quality of intratumoral T cells and thereby response to immunotherapy.
Collapse
Affiliation(s)
- Maria M Steele
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Abhinav Jaiswal
- Department of Dermatology, Microbiology and Immunology, Meyer Cancer Center, Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ines Delclaux
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Ian D Dryg
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Dhaarini Murugan
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Julia Femel
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sunny Son
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY, USA
| | - Haley du Bois
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Cameron Hill
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | - Young H Chang
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, USA
- OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, USA
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Niroshana Anandasabapathy
- Department of Dermatology, Microbiology and Immunology, Meyer Cancer Center, Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
17
|
Heim TA, Lin Z, Steele MM, Mudianto T, Lund AW. CXCR6 promotes dermal CD8 + T cell survival and transition to long-term tissue residence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528487. [PMID: 36824892 PMCID: PMC9949075 DOI: 10.1101/2023.02.14.528487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Tissue resident memory T cells (TRM) provide important protection against infection, and yet the interstitial signals necessary for their formation and persistence remain incompletely understood. Here we show that antigen-dependent induction of the chemokine receptor, CXCR6, is a conserved requirement for TRM formation in peripheral tissue after viral infection. CXCR6 was dispensable for the early accumulation of antigen-specific CD8+ T cells in skin and did not restrain their exit. Single cell sequencing indicated that CXCR6-/- CD8+ T cells were also competent to acquire a transcriptional program of residence but exhibited deficiency in multiple pathways that converged on survival and metabolic signals necessary for memory. As such, CXCR6-/- CD8+ T cells exhibited increased rates of apoptosis relative to controls in the dermis, leading to inefficient TRM formation. CXCR6 expression may therefore represent a common mechanism across peripheral non-lymphoid tissues and inflammatory states that increases the probability of long-term residence.
Collapse
Affiliation(s)
- Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ziyan Lin
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY, USA
| | - Maria M. Steele
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Cell, Developmental & Cancer Biology and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amanda W. Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Cell, Developmental & Cancer Biology and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Brunell AE, Lahesmaa R, Autio A, Thotakura AK. Exhausted T cells hijacking the cancer-immunity cycle: Assets and liabilities. Front Immunol 2023; 14:1151632. [PMID: 37122741 PMCID: PMC10140554 DOI: 10.3389/fimmu.2023.1151632] [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: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
T cell exhaustion is an alternative differentiation path of T cells, sometimes described as a dysfunction. During the last decade, insights of T cell exhaustion acting as a bottle neck in the field of cancer immunotherapy have undoubtedly provoked attention. One of the main drivers of T cell exhaustion is prolonged antigen presentation, a prerequisite in the cancer-immunity cycle. The umbrella term "T cell exhaustion" comprises various stages of T cell functionalities, describing the dynamic, one-way exhaustion process. Together these qualities of T cells at the exhaustion continuum can enable tumor clearance, but if the exhaustion acquired timeframe is exceeded, tumor cells have increased possibilities of escaping immune system surveillance. This could be considered a tipping point where exhausted T cells switch from an asset to a liability. In this review, the contrary role of exhausted T cells is discussed.
Collapse
Affiliation(s)
- Anna E. Brunell
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Anu Autio
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Anil K. Thotakura
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
- *Correspondence: Anil K. Thotakura,
| |
Collapse
|
19
|
Lund AW. Immune Potential Untapped: Leveraging the Lymphatic System for Cancer Immunotherapy. Cancer Immunol Res 2022; 10:1042-1046. [PMID: 35895021 PMCID: PMC9673990 DOI: 10.1158/2326-6066.cir-22-0266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022]
Abstract
Over the past decade, our understanding of the role of the lymphatic vasculature in tumor progression has evolved from it being a passive participant, as a first step along Halsted's path of sequential metastasis, to a potentially active regulator of antitumor immune surveillance. These new data, however, seemingly support paradoxical predictions for cancer immunotherapy; on one hand that enhanced lymphatic involvement augments antitumor immune surveillance and on the other, drives immune evasion and metastasis. The potential to leverage lymphatic biology for the benefit of clinical immunotherapy, therefore, requires a mechanistic understanding of how the lymphatic vasculature interacts with functional immune responses during disease progression and in the context of relevant immunotherapy regimes. In this review, I dissect the promise and challenge of engaging the lymphatic system for therapy and suggest important avenues for future investigation and potential application. See related article, p. 1041.
Collapse
Affiliation(s)
- Amanda W. Lund
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York.,Department of Pathology, NYU Grossman School of Medicine, New York, New York.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
20
|
Sakurai Y, Abe N, Yoshikawa K, Oyama R, Ogasawara S, Murata T, Nakai Y, Tange K, Tanaka H, Akita H. Targeted delivery of lipid nanoparticle to lymphatic endothelial cells via anti-podoplanin antibody. J Control Release 2022; 349:379-387. [PMID: 35787913 DOI: 10.1016/j.jconrel.2022.06.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 12/28/2022]
Abstract
Lymphatic endothelial cells (LECs) that form lymphatic vessels play a pivotal role in immune regulation. It was recently reported that LECs suppress the antigen-dependent anti-tumor immunity in cancer tissues. Thus, regulating the function of LECs is a promising strategy for cancer therapy. The objective of this study was to develop a method for the selective delivery of small interfering RNA (siRNA) to LECs. For this purpose, the siRNA was formulated into nanoparticles (LNPs) to prevent them from being degraded in body fluids and to facilitate their penetration of the cell membrane. A breakthrough technology for achieving this is ONPATTRO®, a world's first siRNA drug. Since LNPs are taken up by hepatocytes relatively well via low-density lipoprotein receptors, most of the LNP systems that have been developed so far target hepatocytes. In this study, we report on the development of a new method for the rapid and convenient method for modifying LNPs with antibodies using the CLick reaction on the Interface of the nanoParticle (CLIP). The CLIP approach was faster and more versatile than the conventional method using amide coupling. As a demonstration, we report on the LEC-targeted siRNA delivery by using antibody-modified LNPs both in vitro and in vivo. The method used for the modification of LNPs is highly promising and has the potential for expanding the LNP-based delivery of nucleic acids in the future.
Collapse
Affiliation(s)
- Yu Sakurai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan.
| | - Nodoka Abe
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Keito Yoshikawa
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Ryotaro Oyama
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Satoshi Ogasawara
- Laboratory of Biostructural Chemistry, Department of Chemistry, Graduate School of Science, Chiba University, Japan; Membrane Protein Research and Molecular Chirality Research Centers, Chiba University, Japan
| | - Takeshi Murata
- Laboratory of Biostructural Chemistry, Department of Chemistry, Graduate School of Science, Chiba University, Japan; Membrane Protein Research and Molecular Chirality Research Centers, Chiba University, Japan
| | - Yuta Nakai
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan
| | - Kota Tange
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan.
| |
Collapse
|
21
|
Doan TA, Forward T, Tamburini BAJ. Trafficking and retention of protein antigens across systems and immune cell types. Cell Mol Life Sci 2022; 79:275. [PMID: 35505125 PMCID: PMC9063628 DOI: 10.1007/s00018-022-04303-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/05/2022]
Abstract
In response to infection or vaccination, the immune system initially responds non-specifically to the foreign insult (innate) and then develops a specific response to the foreign antigen (adaptive). The programming of the immune response is shaped by the dispersal and delivery of antigens. The antigen size, innate immune activation and location of the insult all determine how antigens are handled. In this review we outline which specific cell types are required for antigen trafficking, which processes require active compared to passive transport, the ability of specific cell types to retain antigens and the viruses (human immunodeficiency virus, influenza and Sendai virus, vesicular stomatitis virus, vaccinia virus) and pattern recognition receptor activation that can initiate antigen retention. Both where the protein antigen is localized and how long it remains are critically important in shaping protective immune responses. Therefore, understanding antigen trafficking and retention is necessary to understand the type and magnitude of the immune response and essential for the development of novel vaccine and therapeutic targets.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA.,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, USA. .,Immunology Graduate Program, University of Colorado School of Medicine, Aurora, USA. .,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
22
|
Churchill MJ, du Bois H, Heim TA, Mudianto T, Steele MM, Nolz JC, Lund AW. Infection-induced lymphatic zippering restricts fluid transport and viral dissemination from skin. J Exp Med 2022; 219:e20211830. [PMID: 35353138 PMCID: PMC8972184 DOI: 10.1084/jem.20211830] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 01/19/2022] [Accepted: 03/14/2022] [Indexed: 01/13/2023] Open
Abstract
Lymphatic vessels are often considered passive conduits that flush antigenic material, pathogens, and cells to draining lymph nodes. Recent evidence, however, suggests that lymphatic vessels actively regulate diverse processes from antigen transport to leukocyte trafficking and dietary lipid absorption. Here we tested the hypothesis that infection-induced changes in lymphatic transport actively contribute to innate host defense. We demonstrate that cutaneous vaccinia virus infection by scarification activates dermal lymphatic capillary junction tightening (zippering) and lymph node lymphangiogenesis, which are associated with reduced fluid transport and cutaneous viral sequestration. Lymphatic-specific deletion of VEGFR2 prevented infection-induced lymphatic capillary zippering, increased fluid flux out of tissue, and allowed lymphatic dissemination of virus. Further, a reduction in dendritic cell migration to lymph nodes in the absence of lymphatic VEGFR2 associated with reduced antiviral CD8+ T cell expansion. These data indicate that VEGFR2-driven lymphatic remodeling is a context-dependent, active mechanism of innate host defense that limits viral dissemination and facilitates protective, antiviral CD8+ T cell responses.
Collapse
Affiliation(s)
- Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Haley du Bois
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Maria M. Steele
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Amanda W. Lund
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
23
|
Magold AI, Swartz MA. Pathogenic Exploitation of Lymphatic Vessels. Cells 2022; 11:979. [PMID: 35326430 PMCID: PMC8946894 DOI: 10.3390/cells11060979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Lymphatic vessels provide a critical line of communication between peripheral tissues and their draining lymph nodes, which is necessary for robust immune responses against infectious agents. At the same time, lymphatics help shape the nature and kinetics of immune responses to ensure resolution, limit tissue damage, and prevent autoimmune responses. A variety of pathogens have developed strategies to exploit these functions, from multicellular organisms like nematodes to bacteria, viruses, and prions. While lymphatic vessels serve as transport routes for the dissemination of many pathogens, their hypoxic and immune-suppressive environments can provide survival niches for others. Lymphatics can be exploited as perineural niches, for inter-organ distribution among highly motile carrier cells, as effective replicative niches, and as alternative routes in response to therapy. Recent studies have broadened our understanding of lymphatic involvement in pathogenic spread to include a wider range of pathogens, as well as new mechanisms of exploitation, which we summarize here.
Collapse
Affiliation(s)
- Alexandra I. Magold
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA;
| | - Melody A. Swartz
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA;
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Abstract
Cardiac lymphangiogenesis plays an important physiological role in the regulation of interstitial fluid homeostasis, inflammatory, and immune responses. Impaired or excessive cardiac lymphatic remodeling and insufficient lymph drainage have been implicated in several cardiovascular diseases including atherosclerosis and myocardial infarction (MI). Although the molecular mechanisms underlying the regulation of functional lymphatics are not fully understood, the interplay between lymphangiogenesis and immune regulation has recently been explored in relation to the initiation and development of these diseases. In this field, experimental therapeutic strategies targeting lymphangiogenesis have shown promise by reducing myocardial inflammation, edema and fibrosis, and improving cardiac function. On the other hand, however, whether lymphangiogenesis is beneficial or detrimental to cardiac transplant survival remains controversial. In the light of recent evidence, cardiac lymphangiogenesis, a thriving and challenging field has been summarized and discussed, which may improve our knowledge in the pathogenesis of cardiovascular diseases and transplant biology.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Faculty of Welfare and Health Science, Oita University, Oita, 870-1192, Japan.
| |
Collapse
|
25
|
Leong SP, Pissas A, Scarato M, Gallon F, Pissas MH, Amore M, Wu M, Faries MB, Lund AW. The lymphatic system and sentinel lymph nodes: conduit for cancer metastasis. Clin Exp Metastasis 2021; 39:139-157. [PMID: 34651243 PMCID: PMC8967769 DOI: 10.1007/s10585-021-10123-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/06/2021] [Indexed: 11/03/2022]
Abstract
The lymphatic system is a complicated system consisting of the lymphatic vessels and lymph nodes draining the extracellular fluid containing cellular debris, excess water and toxins to the circulatory system. The lymph nodes serve as a filter, thus, when the lymph fluid returns to the heart, it is completely sterile. In addition, the lymphatic system includes the mucosa-associated lymphoid tissue, such as tonsils, adenoids, Peyers patches in the small bowel and even the appendix. Taking advantage of the drainage system of the lymphatics, cancer cells enter the lymphatic vessels and then the lymph nodes. In general, the lymph nodes may serve as a gateway in the majority of cases in early cancer. Occasionally, the cancer cells may enter the blood vessels. This review article emphasizes the structural integrity of the lymphatic system through which cancer cells may spread. Using melanoma and breast cancer sentinel lymph node model systems, the spread of early cancer through the lymphatic system is progressive in a majority of cases. The lymphatic systems of the internal organs are much more complicated and difficult to study. Knowledge from melanoma and breast cancer spread to the sentinel lymph node may establish the basic principles of cancer metastasis. The goal of this review article is to emphasize the complexity of the lymphatic system. To date, the molecular mechanisms of cancer spread from the cancer microenvironment to the sentinel lymph node and distant sites are still poorly understood and their elucidation should take major priority in cancer metastasis research.
Collapse
Affiliation(s)
- Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA. .,University of California, San Francisco, San Francisco, CA, USA.
| | - Alexander Pissas
- Department of Visceral Surgery General Hospital of Bagnols sur Cèze and of Anatomy Faculty of Medicine of Montpellier, Bagnols sur Ceze, Montpellier, France
| | - Muriel Scarato
- Department of Visceral Surgery General Hospital of Bagnols sur Cèze and of Anatomy Faculty of Medicine of Montpellier, Bagnols sur Ceze, Montpellier, France
| | - Francoise Gallon
- Department of Visceral Surgery General Hospital of Bagnols sur Cèze and of Anatomy Faculty of Medicine of Montpellier, Bagnols sur Ceze, Montpellier, France
| | - Marie Helene Pissas
- Department of Visceral Surgery General Hospital of Bagnols sur Cèze and of Anatomy Faculty of Medicine of Montpellier, Bagnols sur Ceze, Montpellier, France
| | - Miguel Amore
- Vascular Anatomy Lab. III Chair of Anatomy, Faculty of Medicine, Buenos Aires University, Buenos Aires, Argentina.,Phlebology and Lymphology Unit. Cardiovascular Surgery Division, Central Military Hospital, Buenos Aires, Argentina
| | - Max Wu
- California Pacific Medical Center, San Francisco, USA
| | - Mark B Faries
- The Angeles Clinic and Research Institute A Cedars-Sinai Affiliate, Los Angeles, CA, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, Department of Pathology, and NYU Langone Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
26
|
Henderson AR, Ilan IS, Lee E. A bioengineered lymphatic vessel model for studying lymphatic endothelial cell-cell junction and barrier function. Microcirculation 2021; 28:e12730. [PMID: 34569678 DOI: 10.1111/micc.12730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessels (LVs) maintain fluid homeostasis by draining interstitial fluid. A failure in lymphatic drainage triggers lymphatic diseases such as lymphedema. Since lymphatic drainage is regulated by lymphatic barrier function, developing experimental models that assess lymphatic barrier function is critical for better understanding of lymphatic physiology and disease. METHODS We built a lymphatic vessel-on-chip (LV-on-chip) by fabricating a microfluidic device that includes a hollow microchannel embedded in three-dimensional (3D) hydrogel. Employing luminal flow in the microchannel, human lymphatic endothelial cells (LECs) seeded in the microchannel formed an engineered LV exhibiting 3D conduit structure. RESULTS Lymphatic endothelial cells formed relatively permeable junctions in 3D collagen 1. However, adding fibronectin to the collagen 1 apparently tightened LEC junctions. We tested lymphatic barrier function by introducing dextran into LV lumens. While LECs in collagen 1 showed permeable barriers, LECs in fibronectin/collagen 1 showed reduced permeability, which was reversed by integrin α5 inhibition. Mechanistically, LECs expressed inactivated integrin α5 in collagen 1. However, integrin α5 is activated in fibronectin and enhances barrier function. Integrin α5 activation itself also tightened LEC junctions in the absence of fibronectin. CONCLUSIONS Lymphatic vessel-on-chip reveals integrin α5 as a regulator of lymphatic barrier function and provides a platform for studying lymphatic barrier function in various conditions.
Collapse
Affiliation(s)
- Aria R Henderson
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Isabelle S Ilan
- College of Human Ecology, Cornell University, Ithaca, New York, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
27
|
Lucas ED, Schafer JB, Matsuda J, Kraus M, Burchill MA, Tamburini BAJ. PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity. Cell Rep 2021; 33:108258. [PMID: 33053342 PMCID: PMC7688291 DOI: 10.1016/j.celrep.2020.108258] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Although the function of the extracellular region of programmed death ligand 1 (PD-L1) through its interactions with PD-1 on T cells is well studied, little is understood regarding the intracellular domain of PD-L1. Here, we outline a major role for PD-L1 intracellular signaling in the control of dendritic cell (DC) migration from the skin to the draining lymph node (dLN). Using a mutant mouse model, we identify a TSS signaling motif within the intracellular domain of PD-L1. The TSS motif proves critical for chemokine-mediated DC migration to the dLN during inflammation. This loss of DC migration, in the PD-L1 TSS mutant, leads to a significant decline in T cell priming when DC trafficking is required for antigen delivery to the dLN. Finally, the TSS motif is required for chemokine receptor signaling downstream of the Gα subunit of the heterotrimeric G protein complex, ERK phosphorylation, and actin polymerization in DCs. Lucas et al. define three residues within the cytoplasmic tail of PD-L1 that are required for proper dendritic cell migration from the skin to the lymph node. These three-amino-acid residues promote chemokine signaling in dendritic cells and productive T cell responses to skin infections.
Collapse
Affiliation(s)
- Erin D Lucas
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | | | - Madison Kraus
- Gates Summer Research Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA.
| |
Collapse
|
28
|
Abstract
Early engagement of the lymphatic system by solid tumors in peripheral, nonlymphoid tissues is a clinical hallmark of cancer and often forecasts poor prognosis. The significance of lymph node metastasis for distant spread, however, has been questioned by large-scale lymph node dissection trials and the likely prevalence of direct hematogenous metastasis. Still, an emerging appreciation for the immunological role of the tumor-draining lymph node has renewed interest in its basic biology, role in metastatic progression, antitumor immunity, and patient outcomes. In this review, we discuss our current understanding of the early mechanisms through which tumors engage lymphatic transport and condition tumor-draining lymph nodes, the significance of these changes for both metastasis and immunity, and potential implications of the tumor-draining lymph node for immunotherapy.
Collapse
Affiliation(s)
- Haley du Bois
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY 10016
| | - Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY 10016
| | - Amanda W. Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY 10016
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016
- Laura and Isaac Perlmutter Cancer Center NYU Langone Health, New York, NY 10016
| |
Collapse
|
29
|
Stritt S, Koltowska K, Mäkinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med 2021; 27:955-970. [PMID: 34332911 DOI: 10.1016/j.molmed.2021.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.
Collapse
Affiliation(s)
- Simon Stritt
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Katarzyna Koltowska
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden.
| |
Collapse
|
30
|
Steele MM, Lund AW. Afferent Lymphatic Transport and Peripheral Tissue Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 206:264-272. [PMID: 33397740 DOI: 10.4049/jimmunol.2001060] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Lymphatic vessels provide an anatomical framework for immune surveillance and adaptive immune responses. Although appreciated as the route for Ag and dendritic cell transport, peripheral lymphatic vessels are often not considered active players in immune surveillance. Lymphatic vessels, however, integrate contextual cues that directly regulate transport, including changes in intrinsic pumping and capillary remodeling, and express a dynamic repertoire of inflammatory chemokines and adhesion molecules that facilitates leukocyte egress out of inflamed tissue. These mechanisms together contribute to the course of peripheral tissue immunity. In this review, we focus on context-dependent mechanisms that regulate fluid and cellular transport out of peripheral nonlymphoid tissues to provide a framework for understanding the effects of afferent lymphatic transport on immune surveillance, peripheral tissue inflammation, and adaptive immunity.
Collapse
Affiliation(s)
- Maria M Steele
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016; .,Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016; and.,Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
31
|
Sim JH, Ambler WG, Sollohub IF, Howlader MJ, Li TM, Lee HJ, Lu TT. Immune Cell-Stromal Circuitry in Lupus Photosensitivity. THE JOURNAL OF IMMUNOLOGY 2021; 206:302-309. [PMID: 33397744 DOI: 10.4049/jimmunol.2000905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Photosensitivity is a sensitivity to UV radiation (UVR) commonly found in systemic lupus erythematosus (SLE) patients who have cutaneous disease. Upon even ambient UVR exposure, patients can develop inflammatory skin lesions that can reduce the quality of life. Additionally, UVR-exposed skin lesions can be associated with systemic disease flares marked by rising autoantibody titers and worsening kidney disease. Why SLE patients are photosensitive and how skin sensitivity leads to systemic disease flares are not well understood, and treatment options are limited. In recent years, the importance of immune cell-stromal interactions in tissue function and maintenance is being increasingly recognized. In this review, we discuss SLE as an anatomic circuit and review recent findings in the pathogenesis of photosensitivity with a focus on immune cell-stromal circuitry in tissue health and disease.
Collapse
Affiliation(s)
- Ji Hyun Sim
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
| | - William G Ambler
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021
| | - Isabel F Sollohub
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021
| | - Mir J Howlader
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Biochemistry and Structural Biology, Cell Biology, Developmental Biology, and Molecular Biology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065; and
| | - Thomas M Li
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021
| | - Henry J Lee
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10065
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; .,Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065.,Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021
| |
Collapse
|
32
|
Ekeke CN, Russell KL, Joubert K, Bartlett DL, Luketich JD, Soloff AC, Guo ZS, Lotze MT, Dhupar R. Fighting Fire With Fire: Oncolytic Virotherapy for Thoracic Malignancies. Ann Surg Oncol 2021; 28:2715-2727. [PMID: 33575873 PMCID: PMC8043873 DOI: 10.1245/s10434-020-09477-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022]
Abstract
Thoracic malignancies are associated with high mortality rates. Conventional therapy for many of the patients with thoracic malignancies is obviated by a high incidence of locoregional recurrence and distant metastasis. Fortunately, developments in immunotherapy provide effective strategies for both local and systemic treatments that have rapidly advanced during the last decade. One promising approach to cancer immunotherapy is to use oncolytic viruses, which have the advantages of relatively high tumor specificity, selective replication-mediated oncolysis, enhanced antigen presentation, and potential for delivery of immunogenic payloads such as cytokines, with subsequent elicitation of effective antitumor immunity. Several oncolytic viruses including adenovirus, coxsackievirus B3, herpes virus, measles virus, reovirus, and vaccinia virus have been developed and applied to thoracic cancers in preclinical murine studies and clinical trials. This review discusses the current state of oncolytic virotherapy in lung cancer, esophageal cancer, and metastatic malignant pleural effusions and considers its potential as an emergent therapeutic for these patients.
Collapse
Affiliation(s)
- Chigozirim N Ekeke
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kira L Russell
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyla Joubert
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adam C Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zong Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Immunology and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Healthcare System, Surgical Services Division, Pittsburgh, PA, USA.
| |
Collapse
|
33
|
Blair TC, Alice AF, Zebertavage L, Crittenden MR, Gough MJ. The Dynamic Entropy of Tumor Immune Infiltrates: The Impact of Recirculation, Antigen-Specific Interactions, and Retention on T Cells in Tumors. Front Oncol 2021; 11:653625. [PMID: 33968757 PMCID: PMC8101411 DOI: 10.3389/fonc.2021.653625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Analysis of tumor infiltration using conventional methods reveals a snapshot view of lymphocyte interactions with the tumor environment. However, lymphocytes have the unique capacity for continued recirculation, exploring varied tissues for the presence of cognate antigens according to inflammatory triggers and chemokine gradients. We discuss the role of the inflammatory and cellular makeup of the tumor environment, as well as antigen expressed by cancer cells or cross-presented by stromal antigen presenting cells, on recirculation kinetics of T cells. We aim to discuss how current cancer therapies may manipulate lymphocyte recirculation versus retention to impact lymphocyte exclusion in the tumor.
Collapse
Affiliation(s)
- Tiffany C Blair
- Molecular Microbiology and Immunology, Oregon Health and Sciences University (OHSU), Portland, OR, United States.,Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Alejandro F Alice
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Lauren Zebertavage
- Molecular Microbiology and Immunology, Oregon Health and Sciences University (OHSU), Portland, OR, United States.,Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,The Oregon Clinic, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| |
Collapse
|
34
|
Wilson JT, Lund AW. Building new roads to stronger immunity. SCIENCE ADVANCES 2021; 7:eabh3971. [PMID: 33762349 PMCID: PMC11323203 DOI: 10.1126/sciadv.abh3971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
A new cancer vaccine technology builds a network of local lymphatic vessels that paves the way to stronger responses to immunotherapy.
Collapse
Affiliation(s)
- John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA.
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
35
|
Aggio JB, Krmeská V, Ferguson BJ, Wowk PF, Rothfuchs AG. Vaccinia Virus Infection Inhibits Skin Dendritic Cell Migration to the Draining Lymph Node. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:776-784. [PMID: 33419767 PMCID: PMC7851745 DOI: 10.4049/jimmunol.2000928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
There is a paucity of information on dendritic cell (DC) responses to vaccinia virus (VACV), including the traffic of DCs to the draining lymph node (dLN). In this study, using a mouse model of infection, we studied skin DC migration in response to VACV and compared it with the tuberculosis vaccine Mycobacterium bovis bacille Calmette-Guérin (BCG), another live attenuated vaccine administered via the skin. In stark contrast to BCG, skin DCs did not relocate to the dLN in response to VACV. Infection with UV-inactivated VACV or modified VACV Ankara promoted DC movement to the dLN, indicating that interference with skin DC migration requires replication-competent VACV. This suppressive effect of VACV was capable of mitigating responses to a secondary challenge with BCG in the skin, ablating DC migration, reducing BCG transport, and delaying CD4+ T cell priming in the dLN. Expression of inflammatory mediators associated with BCG-triggered DC migration were absent from virus-injected skin, suggesting that other pathways invoke DC movement in response to replication-deficient VACV. Despite adamant suppression of DC migration, VACV was still detected early in the dLN and primed Ag-specific CD4+ T cells. In summary, VACV blocks skin DC mobilization from the site of infection while retaining the ability to access the dLN to prime CD4+ T cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | - Veronika Krmeská
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Brian J Ferguson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Pryscilla Fanini Wowk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | | |
Collapse
|
36
|
Gorgulho CM, Krishnamurthy A, Lanzi A, Galon J, Housseau F, Kaneno R, Lotze MT. Gutting it Out: Developing Effective Immunotherapies for Patients With Colorectal Cancer. J Immunother 2021; 44:49-62. [PMID: 33416261 PMCID: PMC8092416 DOI: 10.1097/cji.0000000000000357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/27/2020] [Indexed: 12/20/2022]
Abstract
Risk factors for colorectal cancer (CRC) include proinflammatory diets, sedentary habits, and obesity, in addition to genetic syndromes that predispose individuals to this disease. Current treatment relies on surgical excision and cytotoxic chemotherapies. There has been a renewed interest in immunotherapy as a treatment option for CRC given the success in melanoma and microsatellite instable (MSI) CRC. Immunotherapy with checkpoint inhibitors only plays a role in the 4%-6% of patients with MSIhigh tumors and even within this subpopulation, response rates can vary from 30% to 50%. Most patients with CRC do not respond to this modality of treatment, even though colorectal tumors are frequently infiltrated with T cells. Tumor cells limit apoptosis and survive following intensive chemotherapy leading to drug resistance and induction of autophagy. Pharmacological or molecular inhibition of autophagy improves the efficacy of cytotoxic chemotherapy in murine models. The microbiome clearly plays an etiologic role, in some or most colon tumors, realized by elegant findings in murine models and now investigated in human clinical trials. Recent results have suggested that cancer vaccines may be beneficial, perhaps best as preventive strategies. The search for therapies that can be combined with current approaches to increase their efficacy, and new knowledge of the biology of CRC are pivotal to improve the care of patients suffering from this disease. Here, we review the basic immunobiology of CRC, current "state-of-the-art" immunotherapies and define those areas with greatest therapeutic promise for the future.
Collapse
Affiliation(s)
- Carolina Mendonça Gorgulho
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, São Paulo State University, UNESP, Botucatu, SP, Brazil
- Department of Pathology, School of Medicine of Botucatu, São Paulo State University, UNESP, Botucatu, SP, Brazil
- DAMP Laboratory, Department of Surgery, University of Pittsburgh, Pittsburgh - PA, USA
| | | | - Anastasia Lanzi
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Franck Housseau
- Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, CRB-I Room 4M59, 1650 Orleans Street, Baltimore, MD, USA
| | - Ramon Kaneno
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, São Paulo State University, UNESP, Botucatu, SP, Brazil
- Department of Pathology, School of Medicine of Botucatu, São Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Michael T. Lotze
- DAMP Laboratory, Department of Surgery, University of Pittsburgh, Pittsburgh - PA, USA
| |
Collapse
|
37
|
Lymphatic Type 1 Interferon Responses Are Critical for Control of Systemic Reovirus Dissemination. J Virol 2021; 95:JVI.02167-20. [PMID: 33208448 PMCID: PMC7851543 DOI: 10.1128/jvi.02167-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
Mammalian orthoreovirus (reovirus) spreads from the site of infection to every organ system in the body via the blood. However, mechanisms that underlie reovirus hematogenous spread remain undefined. Nonstructural protein σ1s is a critical determinant of reovirus bloodstream dissemination that is required for efficient viral replication in many types of cultured cells. Here, we used the specificity of the σ1s protein for promoting hematogenous spread as a platform to uncover a role for lymphatic type 1 interferon (IFN-1) responses in limiting reovirus systemic dissemination. We found that replication of a σ1s-deficient reovirus was restored to wild-type levels in cells with defective interferon-α receptor (IFNAR1) signaling. Reovirus spreads systemically following oral inoculation of neonatal mice, whereas the σ1s-null virus remains localized to the intestine. We found that σ1s enables reovirus spread in the presence of a functional IFN-1 response, as the σ1s-deficient reovirus disseminated comparably to wild-type virus in IFNAR1-/- mice. Lymphatics are hypothesized to mediate reovirus spread from the intestine to the bloodstream. IFNAR1 deletion from cells expressing lymphatic vessel endothelium receptor 1 (LYVE-1), a marker for lymphatic endothelial cells, enabled the σ1s-deficient reovirus to disseminate systemically. Together, our findings indicate that IFN-1 responses in lymphatics limit reovirus dissemination. Our data further suggest that the lymphatics are an important conduit for reovirus hematogenous spread.IMPORTANCE Type 1 interferons (IFN-1) are critical host responses to viral infection. However, the contribution of IFN-1 responses to control of viruses in specific cell and tissue types is not fully defined. Here, we identify IFN-1 responses in lymphatics as important for limiting reovirus dissemination. We found that nonstructural protein σ1s enhances reovirus resistance to IFN-1 responses, as a reovirus mutant lacking σ1s was more sensitive to IFN-1 than wild-type virus. In neonatal mice, σ1s is required for reovirus systemic spread. We used tissue-specific IFNAR1 deletion in combination with the IFN-1-sensitive σ1s-null reovirus as a tool to test how IFN-1 responses in lymphatics affect reovirus systemic spread. Deletion of IFNAR1 in lymphatic cells using Cre-lox technology enabled dissemination of the IFN-1-sensitive σ1s-deficient reovirus. Together, our results indicate that IFN-1 responses in lymphatics are critical for controlling reovirus systemic spread.
Collapse
|
38
|
Wojciechowski S, Virenque A, Vihma M, Galbardi B, Rooney EJ, Keuters MH, Antila S, Koistinaho J, Noe FM. Developmental Dysfunction of the Central Nervous System Lymphatics Modulates the Adaptive Neuro-Immune Response in the Perilesional Cortex in a Mouse Model of Traumatic Brain Injury. Front Immunol 2021; 11:559810. [PMID: 33584640 PMCID: PMC7873607 DOI: 10.3389/fimmu.2020.559810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/26/2020] [Indexed: 01/23/2023] Open
Abstract
Rationale The recently discovered meningeal lymphatic vessels (mLVs) have been proposed to be the missing link between the immune and the central nervous system. The role of mLVs in modulating the neuro-immune response following a traumatic brain injury (TBI), however, has not been analyzed. Parenchymal T lymphocyte infiltration has been previously reported as part of secondary events after TBI, suggestive of an adaptive neuro-immune response. The phenotype of these cells has remained mostly uncharacterized. In this study, we identified subpopulations of T cells infiltrating the perilesional areas 30 days post-injury (an early-chronic time point). Furthermore, we analyzed how the lack of mLVs affects the magnitude and the type of T cell response in the brain after TBI. Methods TBI was induced in K14-VEGFR3-Ig transgenic (TG) mice or in their littermate controls (WT; wild type), applying a controlled cortical impact (CCI). One month after TBI, T cells were isolated from cortical areas ipsilateral or contralateral to the trauma and from the spleen, then characterized by flow cytometry. Lesion size in each animal was evaluated by MRI. Results In both WT and TG-CCI mice, we found a prominent T cell infiltration in the brain confined to the perilesional cortex and hippocampus. The majority of infiltrating T cells were cytotoxic CD8+ expressing a CD44hiCD69+ phenotype, suggesting that these are effector resident memory T cells. K14-VEGFR3-Ig mice showed a significant reduction of infiltrating CD4+ T lymphocytes, suggesting that mLVs could be involved in establishing a proper neuro-immune response. Extension of the lesion (measured as lesion volume from MRI) did not differ between the genotypes. Finally, TBI did not relate to alterations in peripheral circulating T cells, as assessed one month after injury. Conclusions Our results are consistent with the hypothesis that mLVs are involved in the neuro-immune response after TBI. We also defined the resident memory CD8+ T cells as one of the main population activated within the brain after a traumatic injury.
Collapse
Affiliation(s)
- Sara Wojciechowski
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anaïs Virenque
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Maria Vihma
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Barbara Galbardi
- Breast Cancer Unit, Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Erin Jane Rooney
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Meike Hedwig Keuters
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Francesco M. Noe
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
O'Melia MJ, Manspeaker MP, Thomas SN. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunother 2021; 70:2179-2195. [PMID: 33459842 DOI: 10.1007/s00262-020-02792-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/07/2020] [Indexed: 12/21/2022]
Abstract
Triple negative breast cancer (TNBC) is a significant clinical problem to which immunotherapeutic strategies have been applied with limited success. Using the syngeneic E0771 TNBC mouse model, this work explores the potential for antitumor CD8+ T cell immunity to be primed extratumorally in lymphoid tissues and therapeutically leveraged. CD8+ T cell viability and responses within the tumor microenvironment (TME) were found to be severely impaired, effects coincident with local immunosuppression that is recapitulated in lymphoid tissues in late stage disease. Prior to onset of a locally suppressed immune microenvironment, however, CD8+ T cell priming within lymph nodes (LN) that depended on tumor lymphatic drainage remained intact. These results demonstrate tumor-draining LNs (TdLN) to be lymphoid tissue niches that support the survival and antigenic priming of CD8+ T lymphocytes against lymph-draining antigen. The therapeutic effects of and CD8+ T cells response to immune checkpoint blockade were furthermore improved when directed to LNs within the tumor-draining lymphatic basin. Therefore, TdLNs represent a unique potential tumor immunity reservoir in TNBC for which strategies may be developed to improve the effects of ICB immunotherapy.
Collapse
Affiliation(s)
- Meghan J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA. .,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
40
|
Paulson D, Harms R, Ward C, Latterell M, Pazour GJ, Fink DM. Loss of Primary Cilia Protein IFT20 Dysregulates Lymphatic Vessel Patterning in Development and Inflammation. Front Cell Dev Biol 2021; 9:672625. [PMID: 34055805 PMCID: PMC8160126 DOI: 10.3389/fcell.2021.672625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Microenvironmental signals produced during development or inflammation stimulate lymphatic endothelial cells to undergo lymphangiogenesis, in which they sprout, proliferate, and migrate to expand the vascular network. Many cell types detect changes in extracellular conditions via primary cilia, microtubule-based cellular protrusions that house specialized membrane receptors and signaling complexes. Primary cilia are critical for receipt of extracellular cues from both ligand-receptor pathways and physical forces such as fluid shear stress. Here, we report the presence of primary cilia on immortalized mouse and primary adult human dermal lymphatic endothelial cells in vitro and on both luminal and abluminal domains of mouse corneal, skin, and mesenteric lymphatic vessels in vivo. The purpose of this study was to determine the effects of disrupting primary cilia on lymphatic vessel patterning during development and inflammation. Intraflagellar transport protein 20 (IFT20) is part of the transport machinery required for ciliary assembly and function. To disrupt primary ciliary signaling, we generated global and lymphatic endothelium-specific IFT20 knockout mouse models and used immunofluorescence microscopy to quantify changes in lymphatic vessel patterning at E16.5 and in adult suture-mediated corneal lymphangiogenesis. Loss of IFT20 during development resulted in edema, increased and more variable lymphatic vessel caliber and branching, as well as red blood cell-filled lymphatics. We used a corneal suture model to determine ciliation status of lymphatic vessels during acute, recurrent, and tumor-associated inflammatory reactions and wound healing. Primary cilia were present on corneal lymphatics during all of the mechanistically distinct lymphatic patterning events of the model and assembled on lymphatic endothelial cells residing at the limbus, stalk, and vessel tip. Lymphatic-specific deletion of IFT20 cell-autonomously exacerbated acute corneal lymphangiogenesis resulting in increased lymphatic vessel density and branching. These data are the first functional studies of primary cilia on lymphatic endothelial cells and reveal a new dimension in regulation of lymphatic vascular biology.
Collapse
Affiliation(s)
- Delayna Paulson
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Rebecca Harms
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Cody Ward
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Mackenzie Latterell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Darci M. Fink
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
- *Correspondence: Darci M. Fink,
| |
Collapse
|
41
|
Lymph Node Stromal Cells: Mapmakers of T Cell Immunity. Int J Mol Sci 2020; 21:ijms21207785. [PMID: 33096748 PMCID: PMC7588999 DOI: 10.3390/ijms21207785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022] Open
Abstract
Stromal cells (SCs) are strategically positioned in both lymphoid and nonlymphoid organs to provide a scaffold and orchestrate immunity by modulating immune cell maturation, migration and activation. Recent characterizations of SCs have expanded our understanding of their heterogeneity and suggested a functional specialization of distinct SC subsets, further modulated by the microenvironment. Lymph node SCs (LNSCs) have been shown to be particularly important in maintaining immune homeostasis and T cell tolerance. Under inflammation situations, such as viral infections or tumor development, SCs undergo profound changes in their numbers and phenotype and play important roles in contributing to either the activation or the control of T cell immunity. In this review, we highlight the role of SCs located in LNs in shaping peripheral T cell responses in different immune contexts, such as autoimmunity, viral and cancer immunity.
Collapse
|
42
|
Soloff AC, Jones KE, Powers AA, Murthy P, Wang Y, Russell KL, Byrne-Steele M, Lund AW, Yuan JM, Monaco SE, Han J, Dhupar R, Lotze MT. HMGB1 Promotes Myeloid Egress and Limits Lymphatic Clearance of Malignant Pleural Effusions. Front Immunol 2020; 11:2027. [PMID: 33013860 PMCID: PMC7498625 DOI: 10.3389/fimmu.2020.02027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Pleural effusions, when benign, are attributed to cardiac events and suffusion of fluid within the pleural space. When malignant, lymphatic obstruction by tumor and failure to absorb constitutively produced fluid is the predominant formulation. The prevailing view has been challenged recently, namely that the lymphatics are only passive vessels, carrying antigenic fluid to secondary lymphoid sites. Rather, lymphatic vessels can be a selective barrier, efficiently coordinating egress of immune cells and factors within tissues, limiting tumor spread and immune pathology. An alternative explanation, offered here, is that damage associated molecular pattern molecules, released in excess, maintain a local milieu associated with recruitment and retention of immune cells associated with failed lymphatic clearance and functional lymphatic obstruction. We found that levels of high mobility group box 1 (HMGB1) were equally elevated in both benign and malignant pleural effusions (MPEs) and that limited diversity of T cell receptor expressing gamma and delta chain were inversely associated with these levels in MPEs. Acellular fluid from MPEs enhanced γδ T cell proliferation in vitro, while inhibiting cytokine production from γδ T cells and monocytes as well as restricting monocyte chemotaxis. Novel therapeutic strategies, targeting HMGB1 and its neutralization in such effusions as well as direct delivery of immune cells into the pleural space to reconstitute normal physiology should be considered.
Collapse
Affiliation(s)
- Adam C. Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Katherine E. Jones
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amy A. Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Pranav Murthy
- Department of Surgery, Division of Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yue Wang
- Department of Surgery, Division of Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Departments of Immunology and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kira L. Russell
- Department of Surgery, Division of Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Amanda W. Lund
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sara E. Monaco
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jian Han
- iRepertoire, Inc., Huntsville, AL, United States
| | - Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Surgical Services Division, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Michael T. Lotze
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Surgery, Division of Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Departments of Immunology and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
43
|
Role of type I interferons and innate immunity in systemic sclerosis: unbalanced activities on distinct cell types? Curr Opin Rheumatol 2020; 31:569-575. [PMID: 31436583 DOI: 10.1097/bor.0000000000000659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The role of type I IFNs (IFN-I) in the promotion of autoimmunity has been well established. However, its role in the skin fibrosis of systemic sclerosis (SSc) is less clear. IFN-I can participate to tissue repair, and, here, we will consider the extent to which IFN-I's role in SSc skin fibrosis may reflect in part IFN-I functions during wound healing. RECENT FINDINGS Studies are beginning to delineate whether IFN-I has a protective or pathogenic role and how IFN-I affects tissue biology. Recent support for a pathogenic role came from a study depleting plasmacytoid dendritic cells during bleomycin-induced skin fibrosis. The depletion reduced the bleomycin-induced IFN-I-stimulated transcripts and both prevented and reversed fibrosis. Additionally, two recent articles, one identifying SSc endothelial cell injury markers and one showing repressed IFN signaling in SSc keratinocytes, suggest the possibility of unbalanced IFN-I activities on distinct cells types. SUMMARY Recent results support a pathogenic role for IFN-I in skin fibrosis, and recent studies along with others suggest a scenario whereby SSc skin damage results from too much IFN-I-activity driving vasculopathy in combination with too little IFN-I-mediated epidermal integrity and antifibrotic fibroblast phenotype.
Collapse
|
44
|
Abstract
The adaptive immune response is a 500-million-year-old (the "Big Bang" of Immunology) collective set of rearranged and/or selected receptors capable of recognizing soluble and cell surface molecules or shape (B cells, antibody), endogenous and extracellular peptides presented by Major Histocompatibility (MHC) molecules including Class I and Class II (conventional αβ T cells), lipid in the context of MHC-like molecules of the CD1 family (NKT cells), metabolites and B7 family molecules/butyrophilins with stress factors (γδT cells), and stress ligands and absence of MHC molecules (natural killer, NK cells). What makes tumor immunogenic is the recruitment of initially innate immune cells to sites of stress or tissue damage with release of Damage-Associated Molecular Pattern (DAMP) molecules. Subsequent maintenance of a chronic inflammatory state, representing a balance between mature, normalized blood vessels, innate and adaptive immune cells and the tumor provides a complex tumor microenvironment serving as the backdrop for Darwinian selection, tumor elimination, tumor equilibrium, and ultimately tumor escape. Effective immunotherapies are still limited, given the complexities of this highly evolved and selected tumor microenvironment. Cytokine therapies and Immune Checkpoint Blockade (ICB) enable immune effector function and are largely dependent on the shape and size of the B and T cell repertoires (the "adaptome"), now accessible by Next-Generation Sequencing (NGS) and dimer-avoidance multiplexed PCR. How immune effectors access the tumor (infiltrated, immune sequestered, and immune desserts), egress and are organized within the tumor are of contemporary interest and substantial investigation.
Collapse
|
45
|
O'Melia MJ, Lund AW, Thomas SN. The Biophysics of Lymphatic Transport: Engineering Tools and Immunological Consequences. iScience 2019; 22:28-43. [PMID: 31739172 PMCID: PMC6864335 DOI: 10.1016/j.isci.2019.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/25/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
Lymphatic vessels mediate fluid flows that affect antigen distribution and delivery, lymph node stromal remodeling, and cell-cell interactions, to thus regulate immune activation. Here we review the functional role of lymphatic transport and lymph node biomechanics in immunity. We present experimental tools that enable quantitative analysis of lymphatic transport and lymph node dynamics in vitro and in vivo. Finally, we discuss the current understanding for how changes in lymphatic transport and lymph node biomechanics contribute to pathogenesis of conditions including cancer, aging, neurodegeneration, and infection.
Collapse
Affiliation(s)
- Meghan J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Amanda W Lund
- Departments of Cell Developmental Cancer Biology, Molecular Microbiology & Immunology, and Dermatology, Knight Cancer Institute, Oregon Health & Science University, 2720 SW Moody Avenue, KR-CDCB, Portland, OR 97239, USA.
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, 315 Ferst Dr NW, Georgia Institute of Technology, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, 801 Ferst Dr NW, Georgia Institute of Technology, Atlanta, GA 30332, USA; Winship Cancer Institute, 1365 Clifton Rd, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
46
|
Trincot C, Caron KM. Lymphatic Function and Dysfunction in the Context of Sex Differences. ACS Pharmacol Transl Sci 2019; 2:311-324. [PMID: 32259065 PMCID: PMC7089000 DOI: 10.1021/acsptsci.9b00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Indexed: 02/08/2023]
Abstract
Endothelial cells are the building blocks of the blood vascular system and exhibit well-characterized sexually dimorphic phenotypes with regard to chromosomal and hormonal sex, imparting innate genetic and physiological differences between male and female vascular systems and cardiovascular disease. However, even though females are predominantly affected by disorders of lymphatic vascular function, we lack a comprehensive understanding of the effects of sex and sex hormones on lymphatic growth, function, and dysfunction. Here, we attempt to comprehensively evaluate the current understanding of sex as a biological variable influencing lymphatic biology. We first focus on elucidating innate and fundamental differences between the sexes in lymphatic function and development. Next, we delve into lymphatic disease and explore the potential underpinnings toward bias prevalence in the female population. Lastly, we incorporate more broadly the role of the lymphatic system in sex-biased diseases such as cancer, cardiovascular disease, reproductive disorders, and autoimmune diseases to explore whether and how sex differences may influence lymphatic function in the context of these pathologies.
Collapse
Affiliation(s)
- Claire
E. Trincot
- Department of Cell Biology
and Physiology, University of North Carolina
Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building,
CB#7545, Chapel Hill, North
Carolina 27599-7545, United States
| | - Kathleen M. Caron
- Department of Cell Biology
and Physiology, University of North Carolina
Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building,
CB#7545, Chapel Hill, North
Carolina 27599-7545, United States
| |
Collapse
|
47
|
Omura S, Kawai E, Sato F, Martinez NE, Minagar A, Al-Kofahi M, Yun JW, Cvek U, Trutschl M, Alexander JS, Tsunoda I. Theiler's Virus-Mediated Immunopathology in the CNS and Heart: Roles of Organ-Specific Cytokine and Lymphatic Responses. Front Immunol 2018; 9:2870. [PMID: 30619258 PMCID: PMC6295469 DOI: 10.3389/fimmu.2018.02870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/21/2018] [Indexed: 02/05/2023] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) induces different diseases in the central nervous system (CNS) and heart, depending on the mouse strains and time course, with cytokines playing key roles for viral clearance and immune-mediated pathology (immunopathology). In SJL/J mice, TMEV infection causes chronic TMEV-induced demyelinating disease (TMEV-IDD) in the spinal cord about 1 month post-inoculation (p.i.). Unlike other immunopathology models, both pro- and anti-inflammatory cytokines can play dual roles in TMEV-IDD. Pro-inflammatory cytokines play beneficial roles in viral clearance while they are also detrimental in immune-mediated demyelination. Anti-inflammatory cytokines suppress not only protective anti-viral immune responses but also detrimental autoreactive immune responses. Conversely, in C3H mice, TMEV infection induces a non-CNS disease, myocarditis, with three distinctive phases: phase I, viral pathology with interferon and chemokine responses; phase II, immunopathology mediated by acquired immune responses; and phase III, cardiac fibrosis. Although the exact mechanism(s) by which a single virus, TMEV, induces these different diseases in different organs is unclear, our bioinformatics approaches, especially principal component analysis (PCA) of transcriptome data, allow us to identify the key factors contributing to organ-specific immunopathology. The PCA demonstrated that in vitro infection of a cardiomyocyte cell line reproduced the transcriptome profile of phase I in TMEV-induced myocarditis; distinct interferon/chemokine-related responses were induced in vitro in TMEV-infected cardiomyocytes, but not in infected neuronal cells. In addition, the PCA of the in vivo CNS transcriptome data showed that decreased lymphatic marker expressions were weakly associated with inflammation in TMEV infection. Here, dysfunction of lymphatic vessels is shown to potentially contribute to immunopathology by delaying the clearance of cytokines and immune cells from the inflammatory site, although this can also confine the virus at these sites, preventing virus spread via lymphatic vessels. On the other hand, in the heart, dysfunction of lymphatics was associated with reduced lymphatic muscle contractility provoked by pro-inflammatory cytokines. Therefore, TMEV infection may induce different patterns of cytokine expressions as well as lymphatic vessel dysfunction by rather different mechanisms between the CNS and heart, which might explain observed patterns of organ-specific immunopathology.
Collapse
Affiliation(s)
- Seiichi Omura
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan.,Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Eiichiro Kawai
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Fumitaka Sato
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan.,Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Nicholas E Martinez
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahmoud Al-Kofahi
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - J Winny Yun
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Urska Cvek
- Department of Computer Science, Louisiana State University Shreveport, Shreveport, LA, United States
| | - Marjan Trutschl
- Department of Computer Science, Louisiana State University Shreveport, Shreveport, LA, United States
| | - J Steven Alexander
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan.,Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
48
|
Lane RS, Femel J, Breazeale AP, Loo CP, Thibault G, Kaempf A, Mori M, Tsujikawa T, Chang YH, Lund AW. IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J Exp Med 2018; 215:3057-3074. [PMID: 30381467 PMCID: PMC6279400 DOI: 10.1084/jem.20180654] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/16/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022] Open
Abstract
Mechanisms of immune suppression in peripheral tissues counteract protective immunity to prevent immunopathology and are coopted by tumors for immune evasion. While lymphatic vessels facilitate T cell priming, they also exert immune suppressive effects in lymph nodes at steady-state. Therefore, we hypothesized that peripheral lymphatic vessels acquire suppressive mechanisms to limit local effector CD8+ T cell accumulation in murine skin. We demonstrate that nonhematopoietic PD-L1 is largely expressed by lymphatic and blood endothelial cells and limits CD8+ T cell accumulation in tumor microenvironments. IFNγ produced by tissue-infiltrating, antigen-specific CD8+ T cells, which are in close proximity to tumor-associated lymphatic vessels, is sufficient to induce lymphatic vessel PD-L1 expression. Disruption of IFNγ-dependent crosstalk through lymphatic-specific loss of IFNγR boosts T cell accumulation in infected and malignant skin leading to increased viral pathology and tumor control, respectively. Consequently, we identify IFNγR as an immunological switch in lymphatic vessels that balances protective immunity and immunopathology leading to adaptive immune resistance in melanoma.
Collapse
Affiliation(s)
- Ryan S Lane
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
| | - Julia Femel
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
| | - Alec P Breazeale
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
| | - Christopher P Loo
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
| | - Guillaume Thibault
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR
- OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR
| | - Andy Kaempf
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health and Science University, Portland, OR
| | - Motomi Mori
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health and Science University, Portland, OR
| | - Takahiro Tsujikawa
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto City, Kyoto, Japan
| | - Young Hwan Chang
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR
- OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR
| | - Amanda W Lund
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
- Department of Dermatology, Oregon Health and Science University, Portland, OR
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| |
Collapse
|
49
|
Lane RS, Lund AW. Non-hematopoietic Control of Peripheral Tissue T Cell Responses: Implications for Solid Tumors. Front Immunol 2018; 9:2662. [PMID: 30498499 PMCID: PMC6249380 DOI: 10.3389/fimmu.2018.02662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/29/2018] [Indexed: 12/16/2022] Open
Abstract
In response to pathological challenge, the host generates rapid, protective adaptive immune responses while simultaneously maintaining tolerance to self and limiting immune pathology. Peripheral tissues (e.g., skin, gut, lung) are simultaneously the first site of pathogen-encounter and also the location of effector function, and mounting evidence indicates that tissues act as scaffolds to facilitate initiation, maintenance, and resolution of local responses. Just as both effector and memory T cells must adapt to their new interstitial environment upon infiltration, tissues are also remodeled in the context of acute inflammation and disease. In this review, we present the biochemical and biophysical mechanisms by which non-hematopoietic stromal cells and extracellular matrix molecules collaborate to regulate T cell behavior in peripheral tissue. Finally, we discuss how tissue remodeling in the context of tumor microenvironments impairs T cell accumulation and function contributing to immune escape and tumor progression.
Collapse
Affiliation(s)
- Ryan S Lane
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States
| | - Amanda W Lund
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States.,Department of Dermatology, Oregon Health and Science University, Portland, OR, United States.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
50
|
Lucas ED, Finlon JM, Burchill MA, McCarthy MK, Morrison TE, Colpitts TM, Tamburini BAJ. Type 1 IFN and PD-L1 Coordinate Lymphatic Endothelial Cell Expansion and Contraction during an Inflammatory Immune Response. THE JOURNAL OF IMMUNOLOGY 2018; 201:1735-1747. [PMID: 30045970 DOI: 10.4049/jimmunol.1800271] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.
Collapse
Affiliation(s)
- Erin D Lucas
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jeffrey M Finlon
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Matthew A Burchill
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Mary K McCarthy
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tonya M Colpitts
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118; and.,Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|