Skip to main content
Lymphatic Research and Biology logoLink to Lymphatic Research and Biology
. 2013 Sep;11(3):128–135. doi: 10.1089/lrb.2013.0006

Role of Vascular and Lymphatic Endothelial Cells in Hantavirus Pulmonary Syndrome Suggests Targeted Therapeutic Approaches

Erich R Mackow 1,, Elena E Gorbunova 1, Nadine A Dalrymple 1, Irina N Gavrilovskaya 1
PMCID: PMC3780285  PMID: 24024573

Abstract

Background

Hantaviruses in the Americas cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect microvascular and lymphatic endothelial cells and cause dramatic changes in barrier functions without disrupting the endothelium. Hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions. The endothelium of arteries, veins, and lymphatic vessels are unique and central to the function of vast pulmonary capillary beds that regulate pulmonary fluid accumulation.

Results

We have found that HPS-causing hantaviruses alter vascular barrier functions of microvascular and lymphatic endothelial cells by altering receptor and signaling pathway responses that serve to permit fluid tissue influx and clear tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to cause acute pulmonary edema, as well as potential therapeutic targets for reducing the severity of HPS disease.

Conclusions

Here we discuss interactions of HPS-causing hantaviruses with the endothelium, roles for unique lymphatic endothelial responses in HPS, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.

Introduction

The vasculature is constantly exposed to viral pathogens yet only a few viruses specifically target the endothelial cell (EC) lining of vessels and cause acute edematous or hemorrhagic disease. Hantaviruses predominantly infect the endothelial cell lining of vessels and nonlytically cause two diseases: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS).113 The mechanisms by which hantaviruses disrupt fluid barrier integrity and clearance functions of the endothelium are beginning to be disclosed and appear to involve dysregulating EC functions that normally restrict fluid leakage from vessels and clear fluid from tissues.6,1420

Capillaries, veins, and lymphatic vessels are lined by a single layer of ECs that collectively form one of the largest tissues of the body.21,22 The endothelium forms a primary fluid barrier within vessels but serves as more than just a conduit for blood to reach and return from tissues.21,23 The endothelium selectively restricts blood and plasma from entering tissues, regulates immune cell infiltration, and responds to damage by limiting leakage, repairing vessels, and directing angiogenesis.21 These ubiquitous functions require the endothelium to respond to a host of systemic and locally generated factors that alter inter-endothelial cell adherence and fluid barrier properties. Consequently, capillary barrier integrity is redundantly regulated by an array of EC-specific effectors that coordinately balance vascular fluid containment with tissue-specific needs, and respond to a host of systemic and locally generated factors that alter inter-endothelial cell adherens junctions.21,2432 ECs respond to activated platelets and immune cells, clotting cascades, chemokines and cytokines, growth factors, nitric oxide, and hypoxic conditions.21,27,3335 However, ECs also secrete cytokines, complement, and growth factors that positively or negatively impact the adherence and activation of platelets and immune cells, regulate responses to hypoxia, and restrict fluid accumulation in tissues.21,23,25,34,3638 Each of these EC responses is controlled by intertwined sensors and signals aimed at returning the endothelium to a resting state, countering permeabilizing effectors, repairing vessel damage, and restoring fluid and oxygenation levels within tissues.21,24,3944

The unique endothelium of capillaries, veins, and lymphatic vessels is central to their discrete fluid barrier and clearance functions.36,4547 Nonlytic viral infection of microvascular or lymphatic ECs (MECs, LECs) may disengage one or more fluid barrier regulatory mechanisms, thereby increasing vascular leakage or fluid clearance and as a consequence result in tissue edema.4852 However, the accumulation of interstitial fluids can result from either increased endothelial permeability or decreased lymphatic vessel clearance of tissue fluids. Altering LEC responses results in decreased lymphatic vessels clearance functions and lymphedema.36,46,47,53 In the lung, lymphatic vessels clear fluid influx from interstitial spaces and keep pulmonary alveolar spaces relatively dry to permit gas exchange.36,46,47,53 Failure of lymphatic vessels to clear fluids has spawned interest in the role of unique LEC and lymphatic vessel functions and regulation that contribute to edematous disease.

Vascular permeability induced by nonlytic viruses is likely to be multifactorial in nature, resulting from virally altered EC responses and signaling pathways, tissue hypoxia, immune cell and platelet functions, and a collaboration of dysregulated interactions that bypass redundant systems which control normal fluid barrier functions.1417,19,54 Failure of the endothelium to regulate fluid accumulation in tissues has severe pathologic consequences, and during HPS results in localized vascular permeability and acute pulmonary edema that contribute to cardiopulmonary insufficiency and a ∼40% mortality rate.46,9 The mechanisms by which HPS causing hantaviruses induce vascular permeability and acute edema following infection of ECs remains be defined. Recent clues to the role of vascular and lymphatic EC functions suggest potential therapeutic mechanisms that may stabilize the endothelium.

Hantavirus Infection and Disease

Hantaviruses are enveloped, tripartite, negative-sense RNA viruses and the only members of the Bunyaviridae that are directly transmitted to humans by mammalian hosts.13,55,56 The hantavirus genome consists of three segments denoted S, M, and L, based on the length of their RNA segments, respectively.13 The L segment encodes the 250 kDa RNA dependent RNA polymerase.13,55 The S segment encodes a 48 kDa nucleocapsid (N) protein, which is the most abundant hantavirus antigen present in infected cells.13 The M segment encodes two viral surface glycoproteins Gn (64 kDa) and Gc (54 kDa) that are co-translationally cleaved and targeted to the ER/cis-Golgi.13,57 Hantaviruses bud internally into the lumen of the cis-Golgi and exit cells via a secretory mechanism consistent with aberrant vesicular trafficking.13 Hantaviruses are both released from ECs and remain cell-associated through interactions with cell surface receptors.14,54,58 GnGc heterodimers on the virion surface are presumed to bind cellular receptors and mediate viral entry into cells.13,14,19,5764

In vitro, hantaviruses replicate to low titers, with initial viral progeny emerging from infected ECs 18–24 hours post infection (hpi), and ∼5×106 maximal titers days after infection.13 Infection of ECs is nonlytic and the permeability of infected EC monolayers is not increased by infection alone.13,15,65 Prototypic HPS (SNV, ANDV, NY-1V), HFRS (Hantaan virus-HTNV),3,5,10,66,67 and nonpathogenic (Tula virus-TULV and Prospect Hill virus-PHV)6870 hantaviruses all infect human ECs regardless of their ability to cause disease, suggesting that EC entry alone is not a cause of pathogenesis.12,15,71 At least two requirements for hantaviruses to be pathogenic have been determined thus far, the ability of hantaviruses to regulate early interferon responses and the use of specific integrins by pathogenic (ANDV, SNV, NY-1V, PUUV, SEOV, HTNV) but not nonpathogenic (PHV, TULV) hantaviruses.19,61,62,64,7275

At least 17 hantaviruses cause HPS, also termed hantavirus cardiopulmonary syndrome (HCPS). Sin Nombre (SNV) in North America and Andes (ANDV) in South America49,7679 are prototype HPS viruses that cause acute pulmonary edema, cardiopulmonary insufficiency, and ∼35%–40% mortality rates.49,7678,8084 One to two weeks after infection, rapid onset of pulmonary edema and hypoxia occurs 6–12 hours after cough and rapidly progresses in severity.46 Bilateral pulmonary infiltrates may be interstitial or alveolar with large pleural effusions of 2–8 liters at necropsy resulting from pulmonary edema fluid accumulating at up to 1 liter per hour in SNV patients.446,8,9

Hantaviruses are one of only a few viruses that primarily infect the EC lining of the vasculature.8,9,12,71,85 Hantavirus antigen is found predominately in vast pulmonary capillary EC beds but is present in ECs within lymph nodes and throughout the body.8,9,85 However, cytopathic effects are not evident following hantavirus infection of ECs in vitro or in vivo.9,15,65 Histologically, the heart, kidneys, brain, and adrenals are grossly normal, with pulmonary alveoli filled with acellular proteinaceous fluid, yet the alveolar epithelium remains intact.46,8,9 The most striking HPS findings are edematous lungs with up to 8 liters of pleural edema.5,6,8,9 Pulmonary edema fluid contains few leukocytes, is largely serous in nature, and is consistent with the nearly complete loss of an alveolar capillary fluid barrier.46,9 The absence of disrupted endothelium during HPS is similar to edematous pulmonary responses observed in patients with high altitude induced pulmonary edema (HAPE).49,86 The long prodrome and rapid onset of edematous symptoms late after hantavirus infection6 suggests the importance of targeting vessel stability in regulating HPS edema.

Vascular and Lymphatic Endothelium: Control of Vascular Fluid Barrier Functions

The endothelium is a remarkable tissue that functions to meet tissue needs, repair vascular damage, and restrict infection. The endothelium lines a series of discrete vessel types that conduct fluids to and from tissues, direct the transfer of nutrients, wastes, and oxygen, and coordinate tissue responses to changing conditions and pathogens.21,47,52,8789 Vascular ECs serve mainly as a conduit in the lining of high pressure arteries but take on a variety of fluid and cellular barrier functions in low pressure veins and capillaries that innervate organs and tissues.47 Lymphatic vessels have a primary role in draining fluid, proteins, and immune cells from tissues and returning these components to the venous circulation.36,4547 Depending on their location, lymphatic vessels serve discrete fluid barrier and regulatory functions, keeping pulmonary alveolar spaces dry and clearing fluid influx from the lungs.47,90,91 These diverse EC settings require discrete EC functions to effect exchange within large capillary beds of the kidney, liver, and lung.47

The endothelial fluid barrier is primarily derived from unique adherens junctions (AJs) composed of an EC-specific vascular-endothelial cadherin (VE-cadherin).24,28,38,89.9294 EC barrier functions are increased by the presence of cell surface presented VE-cadherin and reduced by the dissociation and internalization of VE-cadherin.24,28,89 Phosphorylated VE-cadherin is internalized by its interaction with intracellular actin complexes, and this process is regulated by a variety of cellular receptors and intracellular signaling pathways. VE-cadherin phosphorylation is downregulated by an EC-specific phosphatase (VE-phosphatase) and several pathways that either directly or indirectly induce AJ assembly and EC integrity by returning VE-cadherin to an unphosphorylated resting state.24,89 Chemokines, cytokines, and growth factors indirectly act on EC adherens junctions to increase vascular permeability and thus have the potential to contribute to pathogenic vascular leakage.27,92

Unique VEGF Receptors Regulate Capillary and Lymphatic Function

Vessel-specific ECs contain unique VEGF receptors 1/2/3 (VEGFR1/2/3) that respond to novel forms of VEGF (VEGF A-E) and positively or negatively impact AJ stability and vascular integrity.91,92,95 VEGF-A binds to EC-specific VEGFR2 receptors and activates a Src-Rac-Pak-VE-cadherin pathway resulting in AJ disassembly and vascular permeability.24,28,89 LECs uniquely express VEGFR3 on their surfaces and respond to VEGF-C/D, but also co-express VEGF-A-responsive VEGFR2 receptors and are further regulated by the formation of VEGFR2/3 heterodimers.33,36,46

A second EC-specific growth factor, angiopoietin-1 (Ang-1), binds to Tie-2 receptors and transdominantly blocks VEGF-A directed permeability. Ang-1 activates an alternate signaling pathway that inhibits VEGFR2 directed Src pathway responses and stabilizes VE-cadherin assembly into AJs. Sphingosine-1-phosphate is a lipid mediator produced by platelets that also stabilizes vessels by engaging Edg-1 receptors on ECs and blocks VEGF-A-directed permeability.15,31,39,43,136,139

VEGF-A was originally discovered as a potent vascular permeability factor that induces acute edema.27,96 VEGF-A reportedly acts within 0.5 mm of its release,97 and circulating soluble VEGFRs prevent VEGF-A from systemically permeabilizing vessels.33,96 VEGF-A is induced by hypoxia, and reduced oxygen levels cause HAPE.34,98,99 This results from activating the hypoxia-induced transcription factor-1a (HIF-1a), which senses oxygen levels and transcriptionally induces VEGF-A.52,100102 VEGF-A further upregulates HIF-1α, forming an autocrine loop which amplifies hypoxia-mediated VEGF- responses and EC permeability during HAPE.52,86,98,103 Although this response fosters increased gas exchange, in continued low oxygen environments, these cellular responses instead cause pulmonary edema and respiratory distress.98,101 These findings indicate that EC responses control capillary leakage through interconnected mechanisms and suggest that altering any number of orchestrated EC barrier functions can result in edema.

Pathogenic Hantavirus Binding to Inactive αvβ3 Integrins Regulates EC Permeability

Several studies demonstrate that monolayers of hantavirus-infected ECs are not permeabilized by infection alone14,15,65 and instead indicate that pathogenic hantavirus infected ECs are hyperpermeabilized by VEGF-A.15 The cellular entry of pathogenic hantaviruses is dependent on the presence of αvβ3 integrins on human ECs, while nonpathogenic hantaviruses PHV and TULV use α5β1 integrins.61,62 Pathogenic hantaviruses bind to inactive basal conformations of αvβ3 integrin receptors on ECs while nonpathogenic hantaviruses interact with discrete integrins.61,62,64 Receptor binding directs viral entry, but at late times post-infection cell-associated hantaviruses also negatively impact αvβ3 integrin functions.1417,19,54 Days after infection, cell-associated pathogenic hantaviruses block αvβ3 integrin directed EC migration and direct the binding of quiescent platelets to the EC surface.14,54

Curiously, αvβ3 integrins present on ECs normally regulate vascular permeability. Ectodomains of αvβ3 and VEGFR2 form complexes that direct EC migration, a process that requires AJ disassembly, yet need to limit VEGF-A induced permeability.92,104 Knocking out β3 integrins or antagonizing αvβ3 results in enhanced VEGF-A directed permeability of capillaries in vivo and in vitro.105107 Inhibiting β3 integrin functions causes vascular permeability disorders.92,107110

Similar to antagonizing or knocking out αvβ3 integrins,92,107 pathogenic hantavirus infection sensitizes ECs to the permeabilizing effects of VEGF-A.15,16 SNV-, ANDV-, and HTNV-infected ECs, but not nonpathogenic PHV or TULV-infected ECs, are hyperresponsive to the permeabilizing effects of VEGF-A,15 and VEGFR2 is hyperphosphorylated following pathogenic hantavirus infection.14,17 Enhanced permeability of infected ECs only occurs days after infection when cell-associated hantaviruses coat the EC surface and inactivate αvβ3 integrins.1416,19,58 These findings, in the context of hypoxic HPS patients, suggest that hantavirus binding to inactive αvβ3 integrins contributes to capillary permeability in HPS. These results further suggest a mechanism for hantavirus-enhanced EC permeability that stems from disrupting normal αvβ3-VEGFR2 interactions and enhanced VEGFR2-Src-VE-cadherin signaling responses that dissociate VE-cadherin from AJs.1416,19,24,92 Collectively, these findings demonstrate that cell surface hantaviruses alter normal EC functions that control VEGF-A directed vascular permeability.1417,54,111

Hantavirus–Endothelial Edemagenic Mechanisms

Prominent pulmonary dysfunction is a component of HPS disease and likely stems from hantavirus infection of ECs that line vast alveolar and renal capillary beds.46,8,9,112,113 Acute pulmonary edema is a hallmark of HPS, and HPS patients arrive at hospitals in acute respiratory distress.4 During HPS, bilateral pulmonary fluid infiltrates accumulate at up to a liter per hour, resulting in respiratory insufficiency and patient hypoxia during a critical phase of the disease.4,6,8,9 The cause of acute edema following hantavirus infection is likely to revolve around the ability of the hantaviruses to infect ECs within alveolar capillary beds that normally regulate edema and gas exchange within the lung. Pathogenic mechanisms accounting for this extraordinary rate of pulmonary fluid accumulation have yet to be defined, but are likely to be multifactorial and require both capillary leakage and decreased pulmonary fluid clearance by lymphatic vessels.6,15,16,114116

Clues to the mechanism of hantavirus-induced edema come from disparate findings on the role of hypoxia in acute pulmonary edema, the role of αvβ3 and VEGFR2 EC responses described above. Recent studies of ANDV-infected LECs further demonstrate that normal EC VEGFR2/3 signaling responses are impaired by pathogenic hantaviruses.6,14,15,19,114 Hypoxia is a prominent component of HPS patients which directs VEGF-A secretion.5,6,8,9 Hypoxia-induced VEGF-A leading to acute pulmonary edema may contribute to both vascular leakage and reduced lymphatic vessel fluid clearance.116 In fact, HPS patient VEGF-A levels were markedly elevated in pulmonary edema fluid and PBMCs in acute early phases of HPS.114 Although a demonstrated role for hypoxia in hantavirus-induced permeability has yet to be conclusively defined, the ability of extracorporal membrane oxygenation (ECMO) to reduce HPS patient mortality4,6 strongly suggests a role for hypoxia and VEGF-A in the acute pulmonary edema of HPS patients.

Potential Role of Hantavirus-Infected LECs in HPS Edema

Pulmonary lymphatic vessels are responsible for clearing fluid from alveoli and providing a semi-dry state that permits gas exchange.45,47 Failure of lymphatic vessels to clear fluids results in lymphedema and suggests an additional mechanism for hantavirus-infected LECs to contribute to acute pulmonary edema during HPS.36,46,47 Analysis of pathology samples from HPS patients indicates that hantavirus antigen is present in LECs of patient lymph nodes.8,9,85 Although less is known about LECs, as described above, LECs express unique cell surface receptors and their integrity is regulated by both VEGF-A and VEGF-C.36,46,47,53 Interestingly, LEC VEGFR3 receptors respond to VEGF-C and are associated with reduced tissue edema,36,53 while inhibiting VEGFR3 signaling results in lymphedema.36

Our recent study indicates that ANDV infects LECs, alters LEC barrier functions, and causes the formation of giant LECs that are likely to alter lymphatic vessel functions.116 However, the role of lymphatic vessels and LEC responses remains to be investigated in HPS patients. ANDV infection of LECs could alter lymphatic vessel responses to VEGF-A/C that contribute to pulmonary edema and HPS. Consistent with this, ANDV-infected LECs resulted in their hyperpermeability in response to VEGF-A that is blocked by VEGF-C. Unexpectedly, it was also found that ∼70% of ANDV-infected LECs were giant cells (viable, 4–5 × normal size). Giant cells are caused by mutations in tuberous sclerosis complex (TSC1/2) proteins resulting in activation of mTOR (mammalian target of rapamycin).117126 Further, VEGFR2 responses activate mTOR, while rapamycin inhibits giant cell and VEGF-A permeability responses.117,119,120,127,128

Following ANDV infection of LECs, both giant cell and permeability responses were inhibited by rapamycin or VEGF-C, suggesting that ANDV alters normal hypoxia-VEGFR2-mTOR signaling pathways of LECs.116 Hypoxia induces VEGF-A/Akt/mTOR signaling but these responses are offset by hypoxia-induced REDD1 expression and the ability of REDD1 to stabilize TSC1/TSC2 complexes and block mTOR activation.117,120,122,124,129132 These findings suggest that ANDV selectively disengages normal hypoxia–VEGF-A responses that control mTOR activation, and instead activate mTOR signaling responses causing giant LECs and LEC dysfunction that contributes to HPS. These findings suggest the potential for targeting pathway specific LEC responses using VEGF-C or rapamycin as a means to regulate ANDV-induced lymphatic dysfunction and reduce pulmonary edema during HPS. The ability of rapamycin and VEGF-C to regulate VEGF-A directed lymphatic vessel responses suggests their potential as HPS therapeutics that may be applicable to additional causes of acute pulmonary edema.

Animal HPS Model

Only ANDV infection of Syrian hamsters (Mesocricetus auratus) serves as a model of hantavirus pathogenesis that mimics human HPS in onset symptoms and lethal acute respiratory disease.18,133,134 Inoculation of Syrian hamsters with ANDV, but not SNV or other HPS causing hantaviruses, induces pathology approximating human disease. ANDV causes a fatal infection of Syrian hamsters with an LD50 of 8 plaque-forming units. The disease is characterized by large pleural effusions, congested lungs, and interstitial pneumonitis in the absence of disrupted endothelium.18,133,134 The onset of pulmonary edema coincides with a rapid increase in viremia on day 6, and large inclusion bodies and vacuoles in ultrastructural studies of infected pulmonary ECs.133,134 Viral antigen was localized to capillary ECs, alveolar macrophages, and splenic follicular marginal zones populated by dendritic cells. Interestingly, depletion of CD4 and CD8 T-cells had no effect on the onset, course, symptoms, or outcome of ANDV infection and indicates the absence of T-cell responses.18 Consistent with the potential involvement of β3 integrins and VEGF-A in this process, ANDV binds to conserved residues within PSI domains of both human and hamster β3 integrins.19,64 The role of lymphatic ECs and responses remain unstudied in the animal model or in HPS patient tissues but may be keys to resolving and reducing the severity of HPS disease. Thus, the mechanism of pathogenesis caused by ANDV is consistent with hypoxia–VEGF-A directed acute pulmonary edema that occurs in the absence of T-cell mediated pathology and may include lymphatic vessel dysfunction.18 Studies in Syrian hamsters provide a means of defining determinants of ANDV pathogenesis and the evaluation of therapeutics that target barrier functions of the vascular and lymphatic endothelium.

Targeted Therapeutic Approaches for Stabilizing the Endothelium

Currently there are no effective therapeutics for hantavirus infections or disease. Antiviral effects of interferon or the nucleoside analog ribavirin are only effective prophylactically or at very early times post-infection.13,135 They appear to target early viral replication but neither is effective 1–2 weeks post-infection after the onset of HPS symptoms.46,135 An alternative approach against viruses with a long disease onset may be to target the acute pathologic response therapeutically instead of viral replication. Since hantaviruses infect and alter fluid barrier functions of the endothelium, targeting EC responses that transiently stabilize the vasculature has the potential to reduce the severity and mortality of HPS.43,136,137 This approach also has the advantage of being implemented at the onset of symptoms where antiviral approaches appear to be ineffective.135

Antibody to VEGFR2 reportedly suppresses VEGF-A induced pulmonary edema and suggests the potential of therapeutically antagonizing VEGFR2-Src-VE-cadherin signaling pathways as a means of reducing acute pulmonary edema during HPS.17,24,43 Several well-studied VEGFR2 and Src inhibitors are in human clinical trials or are used therapeutically to treat human cancers and have the potential to reduce the severity of viral permeability based diseases.17,36,43 In vitro, angiopoietin-1 (Ang-1), sphingosine-1-phosphate (S1P), pazopanib, and dasatinib inhibited EC permeability directed by pathogenic hantaviruses.15,17 VEGFR2-Src signaling inhibitors as well as the S1P analog FTY720 are already in clinical trials or used clinically for other purposes.31,139 The ability of rapamycin to block VEGF-A induced microvascular permeability further suggests its potential as an HPS inhibitor through changes induced in both capillaries and lymphatic vessels.122,128,140 VEGF-C may similarly be used along or in conjunction with other inhibitors to target lymphatic EC dysfunction and enhance fluid clearance from HPS patient tissues.

Targeting EC responses provides a potential means of stabilizing HPS patient vessels and reducing edema. Endothelial cells present a plethora of targets that may regulate virally induced vascular permeability and lymphatic functions. Many compounds targeting pathways which stabilize interendothelial cell adherens junctions are already clinically approved for other indications. Targeting vascular and lymphatic endothelial responses may also be broadly applicable to reducing the severity of a wide range of viral infections that impact the endothelium and cause edematous diseases.

Acknowledgments

We thank Chris Bianco and Eric Roth for technical support for these studies.

Author Disclosure Statement

The authors have no commercial associations relating to the data presented in this manuscript and no conflicts of interest, actual or potential, with information or studies presented.

Studies were supported by NIH, NIAID Grants R01AI47873, PO1AI055621, R21AI1080984, and U54AI57158 (NBC-Lipkin).

References

  • 1.Kanerva M. Mustonen J. Vaheri A. Pathogenesis of puumala and other hantavirus infections. Rev Med Virol. 1998;8:67–86. doi: 10.1002/(sici)1099-1654(199804/06)8:2<67::aid-rmv217>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  • 2.Heyman P. Vaheri A. Lundkvist A, et al. Hantavirus infections in Europe: From virus carriers to a major public-health problem. Expert Rev Anti Infect Ther. 2009;7:205–217. doi: 10.1586/14787210.7.2.205. [DOI] [PubMed] [Google Scholar]
  • 3.Lee H. Lee P. Johnson K. Isolation of the etiologic agent of Korean hemorrhagic fever. J Infect Dis. 1978;137:298–308. doi: 10.1093/infdis/137.3.298. [DOI] [PubMed] [Google Scholar]
  • 4.Chang B, et al. Hantavirus cardiopulmonary syndrome. Semin Respir Crit Care Med. 2007;28:193–200. doi: 10.1055/s-2007-976491. [DOI] [PubMed] [Google Scholar]
  • 5.Duchin JS, et al. Hantavirus pulmonary syndrome: A clinical description of 17 patients with a newly recognized disease. The Hantavirus Study Group. N Engl J Med. 1994;330:949–955. doi: 10.1056/NEJM199404073301401. [DOI] [PubMed] [Google Scholar]
  • 6.Koster F. Mackow E. Pathogenesis of the hantavirus pulmonary syndrome. Future Virol. 2012;7:41–51. [Google Scholar]
  • 7.Nichol ST, et al. Genetic identification of a hantavirus associated with an outbreak of acute respiratory illness. Science. 1993;262:914–917. doi: 10.1126/science.8235615. [DOI] [PubMed] [Google Scholar]
  • 8.Nolte KB, et al. Hantavirus pulmonary syndrome in the United States: A pathological description of a disease caused by a new agent. Human Pathol. 1995;26:110–120. doi: 10.1016/0046-8177(95)90123-x. [DOI] [PubMed] [Google Scholar]
  • 9.Zaki S, et al. Hantavirus pulmonary syndrome: Pathogenesis of an emerging infectious disease. Am J Pathol. 1995;146:552–579. [PMC free article] [PubMed] [Google Scholar]
  • 10.Lahdevirta J, et al. Hantaan virus is related to hemorrhagic fever with renal syndrome in Norway. Lancet. 1982;2:606. doi: 10.1016/s0140-6736(82)90678-x. [DOI] [PubMed] [Google Scholar]
  • 11.Lee HW. Hemorrhagic fever with renal syndrome (HFRS) Scand J Infect Dis Suppl. 1982;36:82–85. [PubMed] [Google Scholar]
  • 12.Yanagihara R. Silverman DJ. Experimental infection of human vascular endothelial cells by pathogenic and nonpathogenic hantaviruses. Arch Virol. 1990;111:281–286. doi: 10.1007/BF01311063. [DOI] [PubMed] [Google Scholar]
  • 13.Schmaljohn C. Lipppincott-Raven; Philadelphia: 2001. Bunyaviridae and their Replication in Virology, Fields, Editor; pp. 1581–1602. [Google Scholar]
  • 14.Gavrilovskaya IN. Gorbunova EE. Mackow ER. Pathogenic hantaviruses direct the adherence of quiescent platelets to infected endothelial cells. J Virol. 2010;84:4832–4839. doi: 10.1128/JVI.02405-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gavrilovskaya IN, et al. Hantaviruses direct endothelial cell permeability by sensitizing cells to the vascular permeability factor VEGF, while angiopoietin 1 and sphingosine 1-phosphate inhibit hantavirus-directed permeability. J Virol. 2008;82:5797–5806. doi: 10.1128/JVI.02397-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gorbunova E. Gavrilovskaya IN. Mackow ER. Pathogenic hantaviruses Andes virus and Hantaan virus induce adherens junction disassembly by directing vascular endothelial cadherin internalization in human endothelial cells. J Virol. 2010;84:7405–7411. doi: 10.1128/JVI.00576-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gorbunova EE, et al. VEGFR2 and Src kinase inhibitors suppress ANDV induced endothelial cell permeability. J Virol. 2011;85:2296–2303. doi: 10.1128/JVI.02319-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hammerbeck CD. Hooper JW. T cells are not required for pathogenesis in the Syrian hamster model of hantavirus pulmonary syndrome. J Virol. 2011;85:9929–9944. doi: 10.1128/JVI.05356-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Raymond T, et al. Pathogenic hantaviruses bind plexin-semaphorin-integrin domains present at the apex of inactive, bent alphavbeta3 integrin conformers. Proc Natl Acad Sci USA. 2005;102:1163–1168. doi: 10.1073/pnas.0406743102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shrivastava-Ranjan P. Rollin PE. Spiropoulou CF. Andes virus disrupts the endothelial cell barrier by induction of vascular endothelial growth factor and downregulation of VE-cadherin. J Virol. 2010;84:11227–11234. doi: 10.1128/JVI.01405-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Aird WC. Endothelium as an organ system. Crit Care Med. 2004;32:S271–279. doi: 10.1097/01.ccm.0000129669.21649.40. [DOI] [PubMed] [Google Scholar]
  • 22.Baumgartner-Parzer SM. Waldhausl WK. The endothelium as a metabolic and endocrine organ: Its relation with insulin resistance. Exp Clin Endocrinol Diabetes. 2001;109:S166–179. doi: 10.1055/s-2001-18579. [DOI] [PubMed] [Google Scholar]
  • 23.Valbuena G. Walke DH. The endothelium as a target for infections. Annu Rev Pathol. 2006;1:171–198. doi: 10.1146/annurev.pathol.1.110304.100031. [DOI] [PubMed] [Google Scholar]
  • 24.Gavard J. Breaking the VE-cadherin bonds. FEBS Lett. 2009;583:1–6. doi: 10.1016/j.febslet.2008.11.032. [DOI] [PubMed] [Google Scholar]
  • 25.Bates DO. Harper SJ. Regulation of vascular permeability by vascular endothelial growth factors. Vascul Pharmacol. 2002;39:225–237. doi: 10.1016/s1537-1891(03)00011-9. [DOI] [PubMed] [Google Scholar]
  • 26.Corada M, et al. Vascular endothelial-cadherin is an important determinant of microvascular integrity in vivo. Proc Natl Acad Sci USA. 1999;96:9815–9820. doi: 10.1073/pnas.96.17.9815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dvorak HF, et al. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol. 1995;146:1029–1039. [PMC free article] [PubMed] [Google Scholar]
  • 28.Gavard J. Gutkind JS. VEGF controls endothelial-cell permeability by promoting the beta-arrestin-dependent endocytosis of VE-cadherin. Nat Cell Biol. 2006;8:1223–1234. doi: 10.1038/ncb1486. [DOI] [PubMed] [Google Scholar]
  • 29.Hippenstiel S, et al. VEGF induces hyperpermeability by a direct action on endothelial cells. Am J Physiol. 1998;274:L678–684. doi: 10.1152/ajplung.1998.274.5.L678. [DOI] [PubMed] [Google Scholar]
  • 30.Lampugnani MG, et al. Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments. J Cell Biol. 2006;174:593–604. doi: 10.1083/jcb.200602080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.McVerry BJ. Garcia JG. Endothelial cell barrier regulation by sphingosine 1-phosphate. J Cell Biochem. 2004;92:1075–1085. doi: 10.1002/jcb.20088. [DOI] [PubMed] [Google Scholar]
  • 32.van Hinsbergh VW. van Nieuw Amerongen GP. Endothelial hyperpermeability in vascular leakage. Vascul Pharmacol. 2002;39:171–172. doi: 10.1016/s1537-1891(03)00006-5. [DOI] [PubMed] [Google Scholar]
  • 33.Breen EC. VEGF in biological control. J Cell Biochem. 2007;102:1358–1367. doi: 10.1002/jcb.21579. [DOI] [PubMed] [Google Scholar]
  • 34.Berger MM, et al. Hypoxia impairs systemic endothelial function in individuals prone to high-altitude pulmonary edema. Am J Respir Crit Care Med. 2005;172:763–767. doi: 10.1164/rccm.200504-654OC. [DOI] [PubMed] [Google Scholar]
  • 35.Coller BS. Shattil SJ. The GPIIb/IIIa (integrin alphaIIbbeta3) odyssey: A technology-driven saga of a receptor with twists, turns, and even a bend. Blood. 2008;112:3011–3025. doi: 10.1182/blood-2008-06-077891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bahram F. Claesson-Welsh L. VEGF-mediated signal transduction in lymphatic endothelial cells. Pathophysiology. 2010;17:253–261. doi: 10.1016/j.pathophys.2009.10.004. [DOI] [PubMed] [Google Scholar]
  • 37.Brakenhielm E. Substrate matters: Reciprocally stimulatory integrin and VEGF signaling in endothelial cells. Circ Res. 2007;101:536–538. doi: 10.1161/CIRCRESAHA.107.160937. [DOI] [PubMed] [Google Scholar]
  • 38.Lampugnani MG. Dejana E. The control of endothelial cell functions by adherens junctions. Novartis Found Symp. 2007;283:4–13. doi: 10.1002/9780470319413.ch2. discussion 13–17, 238–241. [DOI] [PubMed] [Google Scholar]
  • 39.Kobayashi H. Lin PC. Angiopoietin/Tie2 signaling, tumor angiogenesis and inflammatory diseases. Front Biosci. 2005;10:666–674. doi: 10.2741/1561. [DOI] [PubMed] [Google Scholar]
  • 40.Takuwa Y. Takuwa N. Sugimoto N. The Edg family G protein-coupled receptors for lysophospholipids: Their signaling properties and biological activities. J Biochem (Tokyo) 2002;131:767–771. doi: 10.1093/oxfordjournals.jbchem.a003163. [DOI] [PubMed] [Google Scholar]
  • 41.Acevedo LM. Weis SM. Cheresh DA. Robo4 counteracts VEGF signaling. Nat Med. 2008;14:372–373. doi: 10.1038/nm0408-372. [DOI] [PubMed] [Google Scholar]
  • 42.Beauvais DM, et al. Syndecan-1 regulates alphavbeta3 and alphavbeta5 integrin activation during angiogenesis and is blocked by synstatin, a novel peptide inhibitor. J Exp Med. 2009;206:691–705. doi: 10.1084/jem.20081278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gavard J. Patel V. Gutkind JS. Angiopoietin-1 prevents VEGF-induced endothelial permeability by sequestering Src through mDia. Dev Cell. 2008;14:25–36. doi: 10.1016/j.devcel.2007.10.019. [DOI] [PubMed] [Google Scholar]
  • 44.Lampugnani MG. Dejana E. Adherens junctions in endothelial cells regulate vessel maintenance and angiogenesis. Thromb Res. 2007;120:S1–6. doi: 10.1016/S0049-3848(07)70124-X. [DOI] [PubMed] [Google Scholar]
  • 45.Baluk P, et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J Exp Med. 2007;204:2349–2362. doi: 10.1084/jem.20062596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Saharinen P, et al. Lymphatic vasculature: Development, molecular regulation and role in tumor metastasis and inflammation. Trends Immunol. 2004;25:387–395. doi: 10.1016/j.it.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 47.Schraufnagel DE. Lung lymphatic anatomy and correlates. Pathophysiology. 2010;17:337–343. doi: 10.1016/j.pathophys.2009.10.008. [DOI] [PubMed] [Google Scholar]
  • 48.Esser S, et al. Vascular endothelial growth factor induces VE-cadherin tyrosine phosphorylation in endothelial cells. J Cell Sci. 1998;111:1853–1865. doi: 10.1242/jcs.111.13.1853. [DOI] [PubMed] [Google Scholar]
  • 49.Kaner RJ, et al. Lung overexpression of the vascular endothelial growth factor gene induces pulmonary edema. Am J Respir Cell Mol Biol. 2000;22:657–664. doi: 10.1165/ajrcmb.22.6.3779. [DOI] [PubMed] [Google Scholar]
  • 50.Mutlu GM. Sznajder JI. Mechanisms of pulmonary edema clearance. Am J Physiol Lung Cell Mol Physiol. 2005;289:L685–695. doi: 10.1152/ajplung.00247.2005. [DOI] [PubMed] [Google Scholar]
  • 51.Schraufnagel DE, et al. Pulmonary lymphatics and edema accumulation after brief lung injury. Am J Physiol Lung Cell Mol Physiol. 2003;284:L891–897. doi: 10.1152/ajplung.00333.2002. [DOI] [PubMed] [Google Scholar]
  • 52.Dehler M, et al. Hypoxia causes permeability oedema in the constant-pressure perfused rat lung. Eur Respir J. 2006;27:600–606. doi: 10.1183/09031936.06.00061505. [DOI] [PubMed] [Google Scholar]
  • 53.Breslin JW. Yuan SY. Wu MH. VEGF-C alters barrier function of cultured lymphatic endothelial cells through a VEGFR-3-dependent mechanism. Lymphat Res Biol. 2007;5:105–113. doi: 10.1089/lrb.2007.1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gavrilovskaya IN, et al. Pathogenic hantaviruses selectively inhibit beta3 integrin directed endothelial cell migration. Arch Virol. 2002;147:1913–1931. doi: 10.1007/s00705-002-0852-0. [DOI] [PubMed] [Google Scholar]
  • 55.Plyusnin A. Vapalahti O. Vaheri A. Hantaviruses: Genome structure, expression and evolution. J Gen Virol. 1996;77:2677–2687. doi: 10.1099/0022-1317-77-11-2677. [DOI] [PubMed] [Google Scholar]
  • 56.Schmaljohn C. Hjelle B. Hantaviruses: A global disease problem. Emerg Infect Dis. 1997;3:95–104. doi: 10.3201/eid0302.970202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pensiero MN. Hayb J. The Hantaan virus M-segment glycoproteins G1 and G2 can be expressed independently. J Virol. 1992;66:1907–1914. doi: 10.1128/jvi.66.4.1907-1914.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Goldsmith CS, et al. Ultrastructural characteristics of Sin Nombre virus, causative agent of hantavirus pulmonary syndrome. Arch Virol. 1995;140:2107–2122. doi: 10.1007/BF01323234. [DOI] [PubMed] [Google Scholar]
  • 59.Schmaljohn CS. Schmaljohn AL. Dalrymple JM. Hantaan virus M RNA: Coding strategy, nucleotide sequence, and gene order. Virology. 1987;157:31–39. doi: 10.1016/0042-6822(87)90310-2. [DOI] [PubMed] [Google Scholar]
  • 60.Pensiero MN, et al. Expression of the Hantaan virus M genome segment by using a vaccinia virus recombinant. J Virol. 1988;62:696–702. doi: 10.1128/jvi.62.3.696-702.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gavrilovskaya IN, et al. Cellular entry of hantaviruses which cause hemorrhagic fever with renal syndrome is mediated by beta3 integrins. J Virol. 1999;73:3951–3959. doi: 10.1128/jvi.73.5.3951-3959.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gavrilovskaya IN, et al. beta3 Integrins mediate the cellular entry of hantaviruses that cause respiratory failure. Proc Natl Acad Sci USA. 1998;95:7074–7079. doi: 10.1073/pnas.95.12.7074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mackow ER. Gavrilovskaya IN. Cellular receptors and hantavirus pathogenesis. Curr Top Microbiol Immunol. 2001;256:91–115. doi: 10.1007/978-3-642-56753-7_6. [DOI] [PubMed] [Google Scholar]
  • 64.Matthys VS, et al. Andes virus recognition of human and Syrian hamster beta3 integrins is determined by an L33P substitution in the PSI domain. J Virol. 2010;84:352–360. doi: 10.1128/JVI.01013-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sundstrom JB, et al. Hantavirus infection induces the expression of RANTES and IP-10 without causing increased permeability in human lung microvascular endothelial cells. J Virol. 2001;75:6070–6085. doi: 10.1128/JVI.75.13.6070-6085.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Elliott LH, et al. Isolation of the causative agent of hantavirus pulmonary syndrome. Am J Trop Med Hyg. 1994;51:102–108. doi: 10.4269/ajtmh.1994.51.102. [DOI] [PubMed] [Google Scholar]
  • 67.Schmaljohn AL, et al. Isolation and initial characterization of a newfound hantavirus from California. Virology. 1995;206:963–972. doi: 10.1006/viro.1995.1019. [DOI] [PubMed] [Google Scholar]
  • 68.Plyusnin A, et al. Tula virus: A newly detected hantavirus carried by European common voles. J Virol. 1994;68:7833–7839. doi: 10.1128/jvi.68.12.7833-7839.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Vapalahti O, et al. Isolation and characterization of Tula virus, a distinct serotype in the genus Hantavirus, family Bunyaviridae. J Gen Virol. 1996;77:3063–3067. doi: 10.1099/0022-1317-77-12-3063. [DOI] [PubMed] [Google Scholar]
  • 70.Lee PW, et al. Partial characterization of Prospect Hill virus isolated from meadow voles in the United States. J Infect Dis. 1985;152:826–829. doi: 10.1093/infdis/152.4.826. [DOI] [PubMed] [Google Scholar]
  • 71.Pensiero MN, et al. Hantaan virus infection of human endothelial cells. J Virol. 1992;66:5929–5936. doi: 10.1128/jvi.66.10.5929-5936.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Matthys V, et al. The C-terminal 42 residues of the TULV Gn protein regulate interferon induction. J Virol. 2011;85:4752–4760. doi: 10.1128/JVI.01945-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Geimonen E, et al. Pathogenic and nonpathogenic hantaviruses differentially regulate endothelial cell responses. Proc Natl Acad Sci USA. 2002;99:13837–13842. doi: 10.1073/pnas.192298899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Alff PJ, et al. The pathogenic NY-1 hantavirus G1 cytoplasmic tail inhibits RIG-I- and TBK-1-directed interferon responses. J Virol. 2006;80:9676–9686. doi: 10.1128/JVI.00508-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alff PJ, et al. The NY-1 hantavirus Gn cytoplasmic tail coprecipitates TRAF3 and inhibits cellular interferon responses by disrupting TBK1-TRAF3 complex formation. J Virol. 2008;82:9115–9122. doi: 10.1128/JVI.00290-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lopez N, et al. Genetic identification of a new hantavirus causing severe pulmonary syndrome in Argentina. Virology. 1996;220:223–226. doi: 10.1006/viro.1996.0305. [DOI] [PubMed] [Google Scholar]
  • 77.Enria D, et al. Hantavirus pulmonary syndrome in Argentina. Possibility of person to person transmission. Medicina. 1996;56:709–711. [PubMed] [Google Scholar]
  • 78.Padula PJ, et al. Hantavirus pulmonary syndrome outbreak in Argentina: Molecular evidence for person-to-person transmission of Andes virus. Virology. 1998;241:323–330. doi: 10.1006/viro.1997.8976. [DOI] [PubMed] [Google Scholar]
  • 79.Padula PJ, et al. Complete nucleotide sequence of the M RNA segment of Andes virus and analysis of the variability of the termini of the virus S, M and L RNA segments. J Gen Virol. 2002;83:2117–2122. doi: 10.1099/0022-1317-83-9-2117. [DOI] [PubMed] [Google Scholar]
  • 80.Song JW, et al. Isolation of pathogenic hantavirus from white-footed mouse (Peromyscus leucopus) [letter] Lancet. 1994;344:1637. doi: 10.1016/s0140-6736(94)90430-8. [DOI] [PubMed] [Google Scholar]
  • 81.Koster F, et al. Rapid presumptive diagnosis of hantavirus cardiopulmonary syndrome by peripheral blood smear review. Am J Clin Pathol. 2001;116:665–672. doi: 10.1309/CNWF-DC72-QYMR-M8DA. [DOI] [PubMed] [Google Scholar]
  • 82.Levis S, et al. New Hantaviruses causing hantavirus pulmonary syndrome in central America. Lancet. 1997;349:998–999. doi: 10.1016/s0140-6736(05)62895-4. [DOI] [PubMed] [Google Scholar]
  • 83.Bustamante EA. Levy H. Simpson SQ. Pleural fluid characteristics in hantavirus pulmonary syndrome. Chest. 1997;112:1133–1136. doi: 10.1378/chest.112.4.1133. [DOI] [PubMed] [Google Scholar]
  • 84.Hallin GW, et al. Cardiopulmonary manifestations of hantavirus pulmonary syndrome. Crit Care Med. 1996;24:252–258. doi: 10.1097/00003246-199602000-00012. [DOI] [PubMed] [Google Scholar]
  • 85.Cosgriff TM. Lewis RM. Mechanisms of disease in hemorrhagic fever with renal syndrome. Kidney Int Suppl. 1991;35:S72–79. [PubMed] [Google Scholar]
  • 86.Pham I, et al. Hypoxia upregulates VEGF expression in alveolar epithelial cells in vitro and in vivo. Am J Physiol Lung Cell Mol Physiol. 2002;283:L1133–1142. doi: 10.1152/ajplung.00464.2001. [DOI] [PubMed] [Google Scholar]
  • 87.Basu A. Chaturvedi UC. Vascular endothelium: The battlefield of dengue viruses. FEMS Immunol Med Microbiol. 2008;53:287–299. doi: 10.1111/j.1574-695X.2008.00420.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Byzova TV, et al. A mechanism for modulation of cellular responses to VEGF: Activation of the integrins. Mol Cell. 2000;6:851–860. [PubMed] [Google Scholar]
  • 89.Dejana E. Orsenigo F. Lampugnani MG. The role of adherens junctions and VE-cadherin in the control of vascular permeability. J Cell Sci. 2008;121:2115–2122. doi: 10.1242/jcs.017897. [DOI] [PubMed] [Google Scholar]
  • 90.Baluk P, et al. Pathogenesis of persistent lymphatic vessel hyperplasia in chronic airway inflammation. J Clin Invest. 2005;115:247–257. doi: 10.1172/JCI22037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Breslin JW, et al. Vascular endothelial growth factor-C stimulates the lymphatic pump by a VEGF receptor-3-dependent mechanism. Am J Physiol Heart Circ Physiol. 2007;293:H709–718. doi: 10.1152/ajpheart.00102.2007. [DOI] [PubMed] [Google Scholar]
  • 92.Robinson SD, et al. Beta3-integrin regulates vascular endothelial growth factor-A-dependent permeability. Arterioscler Thromb Vasc Biol. 2004;24:2108–2114. doi: 10.1161/01.ATV.0000143857.27408.de. [DOI] [PubMed] [Google Scholar]
  • 93.Nawroth R, et al. VE-PTP and VE-cadherin ectodomains interact to facilitate regulation of phosphorylation and cell contacts. EMBO J. 2002;21:4885–4895. doi: 10.1093/emboj/cdf497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wallez Y. Vilgrain I. Huber P. Angiogenesis: The VE-cadherin switch. Trends Cardiovasc Med. 2006;16:55–59. doi: 10.1016/j.tcm.2005.11.008. [DOI] [PubMed] [Google Scholar]
  • 95.Neufeld G, et al. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9–22. [PubMed] [Google Scholar]
  • 96.Dvorak HF. Discovery of vascular permeability factor (VPF) Exp Cell Res. 2006;312:522–526. doi: 10.1016/j.yexcr.2005.11.026. [DOI] [PubMed] [Google Scholar]
  • 97.Dvorak HF, et al. Distribution of vascular permeability factor (vascular endothelial growth factor) in tumors: Concentration in tumor blood vessels. J Exp Med. 1991;174:1275–1278. doi: 10.1084/jem.174.5.1275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Christou H, et al. Increased vascular endothelial growth factor production in the lungs of rats with hypoxia-induced pulmonary hypertension. Am J Respir Cell Mol Biol. 1998;18:768–776. doi: 10.1165/ajrcmb.18.6.2980. [DOI] [PubMed] [Google Scholar]
  • 99.Mukhopadhyay D, et al. Hypoxic induction of human vascular endothelial growth factor expression through c-Src activation. Nature. 1995;375:577–581. doi: 10.1038/375577a0. [DOI] [PubMed] [Google Scholar]
  • 100.Holmes K, et al. Vascular endothelial growth factor receptor-2: Structure, function, intracellular signalling and therapeutic inhibition. Cell Signal. 2007;19:2003–2012. doi: 10.1016/j.cellsig.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 101.Stenmark KR. Fagan KA. Frid MG. Hypoxia-induced pulmonary vascular remodeling: Cellular and molecular mechanisms. Circ Res. 2006;99:675–691. doi: 10.1161/01.RES.0000243584.45145.3f. [DOI] [PubMed] [Google Scholar]
  • 102.Tang N, et al. Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell. 2004;6:485–495. doi: 10.1016/j.ccr.2004.09.026. [DOI] [PubMed] [Google Scholar]
  • 103.Sidel'nikov Iu N. Anisimova NI. [The mechanisms of the development of tissue hypoxia and the criteria for its assessment in patients with hemorrhagic fever with renal syndrome] Ter Arkh. 1991;63:68–70. [PubMed] [Google Scholar]
  • 104.Borges E. Jan Y. Ruoslahti E. Platelet-derived growth factor receptor beta and vascular endothelial growth factor receptor 2 bind to the beta 3 integrin through its extracellular domain. J Biol Chem. 2000;275:39867–39873. doi: 10.1074/jbc.M007040200. [DOI] [PubMed] [Google Scholar]
  • 105.Soker S, et al. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998;92:735–745. doi: 10.1016/s0092-8674(00)81402-6. [DOI] [PubMed] [Google Scholar]
  • 106.Wang L, et al. Neuropilin-1-mediated vascular permeability factor/vascular endothelial growth factor-dependent endothelial cell migration. J Biol Chem. 2003;278:48848–48860. doi: 10.1074/jbc.M310047200. [DOI] [PubMed] [Google Scholar]
  • 107.Weis SM. Cheresh DA. alphav Integrins in angiogenesis and cancer. Cold Spring Harb Perspect Med. 2012;1:a006478. doi: 10.1101/cshperspect.a006478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hodivala-Dilke KM, et al. Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival. J Clin Invest. 1999;103:229–238. doi: 10.1172/JCI5487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hynes RO. Bader BL. Hodivala-Dilke K. Integrins in vascular development [In Process Citation] Braz J Med Biol Res. 1999;32:501–510. doi: 10.1590/s0100-879x1999000500002. [DOI] [PubMed] [Google Scholar]
  • 110.Hynes RO. Structural biology. Changing partners. Science. 2003;300:755–756. doi: 10.1126/science.1084854. [DOI] [PubMed] [Google Scholar]
  • 111.Pepini T, et al. Andes virus regulation of cellular microRNAs contributes to hantavirus induced endothelial cell permeability. J Virol. 2010;84:11929–11936. doi: 10.1128/JVI.01658-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lahdevirta J. Clinical features of HFRS in Scandinavia as compared with East Asia. Scand J Infect Dis Suppl. 1982;36:93–95. [PubMed] [Google Scholar]
  • 113.Khan AS, et al. Hantavirus pulmonary syndrome: The first 100 US cases. J Infect Dis. 1996;173:1297–1303. doi: 10.1093/infdis/173.6.1297. [DOI] [PubMed] [Google Scholar]
  • 114.Gavrilovskaya I, et al. Elevated VEGF levels in pulmonary edema fluid and PBMCs from patients with acute hantavirus pulmonary syndrome. Adv Virol. 2012;2012:674360. doi: 10.1155/2012/674360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gavrilovskaya I, et al. The role of the endothelium in HPS pathogenesis and potential therapeutic approaches. Adv Virol. 2012;2012:467059. doi: 10.1155/2012/467059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gavrilovskaya IN. Gorbunova EE. Mackow ER. Andes virus infection of lymphatic endothelial cells causes giant cell and enhanced permeability responses that are rapamycin and vascular endothelial growth factor C sensitive. J Virol. 2012;86:8765–8772. doi: 10.1128/JVI.00817-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.El-Hashemite N, et al. Loss of Tsc1 or Tsc2 induces vascular endothelial growth factor production through mammalian target of rapamycin. Cancer Res. 2003;63:5173–5177. [PubMed] [Google Scholar]
  • 118.Hay N. Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18:1926–1945. doi: 10.1101/gad.1212704. [DOI] [PubMed] [Google Scholar]
  • 119.Major P. Potential of mTOR inhibitors for the treatment of subependymal giant cell astrocytomas in tuberous sclerosis complex. Aging (Albany NY) 2011;3:189–191. doi: 10.18632/aging.100298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Orlova KA. Crino PB. The tuberous sclerosis complex. Ann NY Acad Sci. 2010;1184:87–105. doi: 10.1111/j.1749-6632.2009.05117.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kobayashi S, et al. Rapamycin, a specific inhibitor of the mammalian target of rapamycin, suppresses lymphangiogenesis and lymphatic metastasis. Cancer Sci. 2007;98:726–733. doi: 10.1111/j.1349-7006.2007.00439.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Land SC. Tee AR. Hypoxia-inducible factor 1alpha is regulated by the mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J Biol Chem. 2007;282:20534–20543. doi: 10.1074/jbc.M611782200. [DOI] [PubMed] [Google Scholar]
  • 123.Xue Q, et al. Rapamycin inhibition of the Akt/mTOR pathway blocks select stages of VEGF-A164-driven angiogenesis, in part by blocking S6Kinase. Arterioscler Thromb Vasc Biol. 2009;29:1172–1178. doi: 10.1161/ATVBAHA.109.185918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Brugarolas J, et al. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 2004;18:2893–2904. doi: 10.1101/gad.1256804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Brugarolas JB, et al. TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell. 2003;4:147–158. doi: 10.1016/s1535-6108(03)00187-9. [DOI] [PubMed] [Google Scholar]
  • 126.Brugarolas J. Kaelin WG., Jr. Dysregulation of HIF and VEGF is a unifying feature of the familial hamartoma syndromes. Cancer Cell. 2004;6:7–10. doi: 10.1016/j.ccr.2004.06.020. [DOI] [PubMed] [Google Scholar]
  • 127.Humar R, et al. Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)-dependent signaling. FASEB J. 2002;16:771–780. doi: 10.1096/fj.01-0658com. [DOI] [PubMed] [Google Scholar]
  • 128.Kim DD, et al. Rapamycin inhibits VEGF-induced microvascular hyperpermeability in vivo. Microcirculation. 2009;17:128–136. doi: 10.1111/j.1549-8719.2009.00012.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Laplante M. Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274–293. doi: 10.1016/j.cell.2012.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Regazzetti C, et al. Insulin induces REDD1 expression through hypoxia-inducible factor 1 activation in adipocytes. J Biol Chem. 2009;285:5157–564. doi: 10.1074/jbc.M109.047688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Regazzetti C, et al. Regulated in development, DNA damage responses −1 (REDD1) protein contributes to insulin signaling pathway in adipocytes. PLoS One. 2012;7:e52154. doi: 10.1371/journal.pone.0052154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Ruvinsky I. Meyuhas O. Ribosomal protein S6 phosphorylation: From protein synthesis to cell size. Trends Biochem Sci. 31:342–348. doi: 10.1016/j.tibs.2006.04.003. 200. [DOI] [PubMed] [Google Scholar]
  • 133.Hooper JW, et al. A lethal disease model for hantavirus pulmonary syndrome. Virology. 2001;289:6–14. doi: 10.1006/viro.2001.1133. [DOI] [PubMed] [Google Scholar]
  • 134.Wahl-Jensen V, et al. Temporal analysis of Andes virus and Sin Nombre virus infections of Syrian hamsters. J Virol. 2007;81:7449–7462. doi: 10.1128/JVI.00238-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Jonsson CB. Hooper J. Mertz G. Treatment of hantavirus pulmonary syndrome. Antiviral Res. 2008;78:162–169. doi: 10.1016/j.antiviral.2007.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Garcia JG, et al. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest. 2001;108:689–701. doi: 10.1172/JCI12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Jones CA, et al. Robo4 stabilizes the vascular network by inhibiting pathologic angiogenesis and endothelial hyperpermeability. Nat Med. 2008;14:448–453. doi: 10.1038/nm1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Acevedo LM, et al. Semaphorin 3A suppresses VEGF-mediated angiogenesis yet acts as a vascular permeability factor. Blood. 2008;111:2674–2680. doi: 10.1182/blood-2007-08-110205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.McVerry BJ, et al. Sphingosine 1-phosphate reduces vascular leak in murine and canine models of acute lung injury. Am J Respir Crit Care Med. 2004;170:987–993. doi: 10.1164/rccm.200405-684OC. [DOI] [PubMed] [Google Scholar]
  • 140.Huber S, et al. Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis. Kidney Int. 2007;71:771–777. doi: 10.1038/sj.ki.5002112. [DOI] [PubMed] [Google Scholar]

Articles from Lymphatic Research and Biology are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES