Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38659941

ABSTRACT

In search for broad-spectrum antivirals, we discovered a small molecule inhibitor, RMC-113, that potently suppresses the replication of multiple RNA viruses including SARS-CoV-2 in human lung organoids. We demonstrated selective dual inhibition of the lipid kinases PIP4K2C and PIKfyve by RMC-113 and target engagement by its clickable analog. Advanced lipidomics revealed alteration of SARS-CoV-2-induced phosphoinositide signature by RMC-113 and linked its antiviral effect with functional PIP4K2C and PIKfyve inhibition. We discovered PIP4K2C's roles in SARS-CoV-2 entry, RNA replication, and assembly/egress, validating it as a druggable antiviral target. Integrating proteomics, single-cell transcriptomics, and functional assays revealed that PIP4K2C binds SARS-CoV-2 nonstructural protein 6 and regulates virus-induced impairment of autophagic flux. Reversing this autophagic flux impairment is a mechanism of antiviral action of RMC-113. These findings reveal virus-induced autophagy regulation via PIP4K2C, an understudied kinase, and propose dual inhibition of PIP4K2C and PIKfyve as a candidate strategy to combat emerging viruses.

2.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37581931

ABSTRACT

Targeting host factors exploited by multiple viruses could offer broad-spectrum solutions for pandemic preparedness. Seventeen candidates targeting diverse functions emerged in a screen of 4,413 compounds for SARS-CoV-2 inhibitors. We demonstrated that lapatinib and other approved inhibitors of the ErbB family of receptor tyrosine kinases suppress replication of SARS-CoV-2, Venezuelan equine encephalitis virus (VEEV), and other emerging viruses with a high barrier to resistance. Lapatinib suppressed SARS-CoV-2 entry and later stages of the viral life cycle and showed synergistic effect with the direct-acting antiviral nirmatrelvir. We discovered that ErbB1, ErbB2, and ErbB4 bind SARS-CoV-2 S1 protein and regulate viral and ACE2 internalization, and they are required for VEEV infection. In human lung organoids, lapatinib protected from SARS-CoV-2-induced activation of ErbB-regulated pathways implicated in non-infectious lung injury, proinflammatory cytokine production, and epithelial barrier injury. Lapatinib suppressed VEEV replication, cytokine production, and disruption of blood-brain barrier integrity in microfluidics-based human neurovascular units, and reduced mortality in a lethal infection murine model. We validated lapatinib-mediated inhibition of ErbB activity as an important mechanism of antiviral action. These findings reveal regulation of viral replication, inflammation, and tissue injury via ErbBs and establish a proof of principle for a repurposed, ErbB-targeted approach to combat emerging viruses.


Subject(s)
COVID-19 , Hepatitis C, Chronic , Animals , Humans , Mice , Antiviral Agents/pharmacology , Cytokines , Inflammation/drug therapy , Lapatinib/pharmacology , SARS-CoV-2
3.
bioRxiv ; 2023 May 10.
Article in English | MEDLINE | ID: mdl-34159337

ABSTRACT

Targeting host factors exploited by multiple viruses could offer broad-spectrum solutions for pandemic preparedness. Seventeen candidates targeting diverse functions emerged in a screen of 4,413 compounds for SARS-CoV-2 inhibitors. We demonstrated that lapatinib and other approved inhibitors of the ErbB family receptor tyrosine kinases suppress replication of SARS-CoV-2, Venezuelan equine encephalitis virus (VEEV), and other emerging viruses with a high barrier to resistance. Lapatinib suppressed SARS-CoV-2 entry and later stages of the viral life cycle and showed synergistic effect with the direct-acting antiviral nirmatrelvir. We discovered that ErbB1, 2 and 4 bind SARS-CoV-2 S1 protein and regulate viral and ACE2 internalization, and they are required for VEEV infection. In human lung organoids, lapatinib protected from SARS-CoV-2-induced activation of ErbB-regulated pathways implicated in non-infectious lung injury, pro-inflammatory cytokine production, and epithelial barrier injury. Lapatinib suppressed VEEV replication, cytokine production and disruption of the blood-brain barrier integrity in microfluidic-based human neurovascular units, and reduced mortality in a lethal infection murine model. We validated lapatinib-mediated inhibition of ErbB activity as an important mechanism of antiviral action. These findings reveal regulation of viral replication, inflammation, and tissue injury via ErbBs and establish a proof-of-principle for a repurposed, ErbB-targeted approach to combat emerging viruses.

4.
Front Immunol ; 13: 899617, 2022.
Article in English | MEDLINE | ID: mdl-35720389

ABSTRACT

COVID-19 emergency use authorizations and approvals for vaccines were achieved in record time. However, there remains a need to develop additional safe, effective, easy-to-produce, and inexpensive prevention to reduce the risk of acquiring SARS-CoV-2 infection. This need is due to difficulties in vaccine manufacturing and distribution, vaccine hesitancy, and, critically, the increased prevalence of SARS-CoV-2 variants with greater contagiousness or reduced sensitivity to immunity. Antibodies from eggs of hens (immunoglobulin Y; IgY) that were administered the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein were developed for use as nasal drops to capture the virus on the nasal mucosa. Although initially raised against the 2019 novel coronavirus index strain (2019-nCoV), these anti-SARS-CoV-2 RBD IgY surprisingly had indistinguishable enzyme-linked immunosorbent assay binding against variants of concern that have emerged, including Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529). This is different from sera of immunized or convalescent patients. Culture neutralization titers against available Alpha, Beta, and Delta were also indistinguishable from the index SARS-CoV-2 strain. Efforts to develop these IgY for clinical use demonstrated that the intranasal anti-SARS-CoV-2 RBD IgY preparation showed no binding (cross-reactivity) to a variety of human tissues and had an excellent safety profile in rats following 28-day intranasal delivery of the formulated IgY. A double-blind, randomized, placebo-controlled phase 1 study evaluating single-ascending and multiple doses of anti-SARS-CoV-2 RBD IgY administered intranasally for 14 days in 48 healthy adults also demonstrated an excellent safety and tolerability profile, and no evidence of systemic absorption. As these antiviral IgY have broad selectivity against many variants of concern, are fast to produce, and are a low-cost product, their use as prophylaxis to reduce SARS-CoV-2 viral transmission warrants further evaluation. Clinical Trial Registration: https://www.clinicaltrials.gov/ct2/show/NCT04567810, identifier NCT04567810.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Viral , COVID-19/prevention & control , Chickens , Female , Humans , Immunoglobulins , Rats , Spike Glycoprotein, Coronavirus
5.
Cell Rep ; 39(7): 110812, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35568025

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-neutralizing monoclonal antibodies (mAbs) can reduce the risk of hospitalization from coronavirus disease 2019 (COVID-19) when administered early. However, SARS-CoV-2 variants of concern (VOCs) have negatively affected therapeutic use of some authorized mAbs. Using a high-throughput B cell screening pipeline, we isolated LY-CoV1404 (bebtelovimab), a highly potent SARS-CoV-2 spike glycoprotein receptor binding domain (RBD)-specific antibody. LY-CoV1404 potently neutralizes authentic SARS-CoV-2, B.1.1.7, B.1.351, and B.1.617.2. In pseudovirus neutralization studies, LY-CoV1404 potently neutralizes variants, including B.1.1.7, B.1.351, B.1.617.2, B.1.427/B.1.429, P.1, B.1.526, B.1.1.529, and the BA.2 subvariant. Structural analysis reveals that the contact residues of the LY-CoV1404 epitope are highly conserved, except for N439 and N501. The binding and neutralizing activity of LY-CoV1404 is unaffected by the most common mutations at these positions (N439K and N501Y). The broad and potent neutralization activity and the relatively conserved epitope suggest that LY-CoV1404 has the potential to be an effective therapeutic agent to treat all known variants.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral , Epitopes , Humans
6.
Viruses ; 14(2)2022 02 10.
Article in English | MEDLINE | ID: mdl-35215962

ABSTRACT

Understanding the magnitude of responses to vaccination during the ongoing SARS-CoV-2 pandemic is essential for ultimate mitigation of the disease. Here, we describe a cohort of 102 subjects (70 COVID-19-naïve, 32 COVID-19-experienced) who received two doses of one of the mRNA vaccines (BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna)). We document that a single exposure to antigen via infection or vaccination induces a variable antibody response which is affected by age, gender, race, and co-morbidities. In response to a second antigen dose, both COVID-19-naïve and experienced subjects exhibited elevated levels of anti-spike and SARS-CoV-2 neutralizing activity; however, COVID-19-experienced individuals achieved higher antibody levels and neutralization activity as a group. The COVID-19-experienced subjects exhibited no significant increase in antibody or neutralization titer in response to the second vaccine dose (i.e., third antigen exposure). Finally, we found that COVID-19-naïve individuals who received the Moderna vaccine exhibited a more robust boost response to the second vaccine dose (p = 0.004) as compared to the response to Pfizer-BioNTech. Ongoing studies with this cohort will continue to contribute to our understanding of the range and durability of responses to SARS-CoV-2 mRNA vaccines.


Subject(s)
2019-nCoV Vaccine mRNA-1273/immunology , Antibodies, Viral/blood , BNT162 Vaccine/immunology , COVID-19/prevention & control , Immunogenicity, Vaccine , SARS-CoV-2/immunology , Vaccination/statistics & numerical data , 2019-nCoV Vaccine mRNA-1273/administration & dosage , Adult , Antibodies, Viral/immunology , Antibody Formation , BNT162 Vaccine/administration & dosage , COVID-19/immunology , Cohort Studies , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged
7.
bioRxiv ; 2022 Mar 24.
Article in English | MEDLINE | ID: mdl-33972947

ABSTRACT

SARS-CoV-2 neutralizing monoclonal antibodies (mAbs) can reduce the risk of hospitalization when administered early during COVID-19 disease. However, the emergence of variants of concern has negatively impacted the therapeutic use of some authorized mAbs. Using a high throughput B-cell screening pipeline, we isolated a highly potent SARS-CoV-2 spike glycoprotein receptor binding domain (RBD)-specific antibody called LY-CoV1404 (also known as bebtelovimab). LY-CoV1404 potently neutralizes authentic SARS-CoV-2 virus, including the prototype, B.1.1.7, B.1.351 and B.1.617.2). In pseudovirus neutralization studies, LY-CoV1404 retains potent neutralizing activity against numerous variants including B.1.1.7, B.1.351, B.1.617.2, B.1.427/B.1.429, P.1, B.1.526, B.1.1.529, and the BA.2 subvariant and retains binding to spike proteins with a variety of underlying RBD mutations including K417N, L452R, E484K, and N501Y. Structural analysis reveals that the contact residues of the LY-CoV1404 epitope are highly conserved with the exception of N439 and N501. Notably, the binding and neutralizing activity of LY-CoV1404 is unaffected by the most common mutations at these positions (N439K and N501Y). The breadth of reactivity to amino acid substitutions present among current VOC together with broad and potent neutralizing activity and the relatively conserved epitope suggest that LY-CoV1404 has the potential to be an effective therapeutic agent to treat all known variants causing COVID-19. In Brief: LY-CoV1404 is a potent SARS-CoV-2-binding antibody that neutralizes all known variants of concern and whose epitope is rarely mutated. Highlights: LY-CoV1404 potently neutralizes SARS-CoV-2 authentic virus and known variants of concern including the B.1.1.529 (Omicron), the BA.2 Omicron subvariant, and B.1.617.2 (Delta) variantsNo loss of potency against currently circulating variantsBinding epitope on RBD of SARS-CoV-2 is rarely mutated in GISAID databaseBreadth of neutralizing activity and potency supports clinical development.

8.
Virulence ; 12(1): 430-443, 2021 12.
Article in English | MEDLINE | ID: mdl-33487119

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus that can cause debilitating, acute febrile illness and potentially result in encephalitis. Currently, there are no FDA-licensed vaccines or specific therapeutics for VEEV. Previous studies have demonstrated that VEEV infection results in increased blood-brain barrier (BBB) permeability that is mediated by matrix metalloproteinases (MMPs). Furthermore, after subarachnoid hemorrhage in mice, MMP-9 is upregulated in the brain and mediates BBB permeability in a toll-like receptor 4 (TLR4)-dependent manner. Here, we demonstrate that disease in C3H mice during VEEV TC-83 infection is dependent on TLR4 because intranasal infection of C3H/HeN (TLR4 WT ) mice with VEEV TC-83 resulted in mortality as opposed to survival of TLR4-defective C3H/HeJ (TLR4 mut ) mice. In addition, BBB permeability was induced to a lesser extent in TLR4 mut mice compared with TLR4 WT mice during VEEV TC-83 infection as determined by sodium fluorescein and fluorescently-conjugated dextran extravasation. Moreover, MMP-9, MMP-2, ICAM-1, CCL2 and IFN-γ were all induced to significantly lower levels in the brains of infected TLR4 mut mice compared with infected TLR4 WT mice despite the absence of significantly different viral titers or immune cell populations in the brains of infected TLR4 WT and TLR4 mut mice. These data demonstrate the critical role of TLR4 in mediating BBB permeability and disease in C3H mice during VEEV TC-83 infection, which suggests that TLR4 is a potential target for the development of therapeutics for VEEV.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalitis Virus, Venezuelan Equine/pathogenicity , Toll-Like Receptor 4/genetics , Animals , Brain/virology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/virology , Female , Mice , Mice, Inbred C3H , Permeability , Toll-Like Receptor 4/metabolism , Virus Replication
9.
PLoS Pathog ; 15(11): e1008061, 2019 11.
Article in English | MEDLINE | ID: mdl-31697791

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes persistent arthritis in a subset of human patients. We report the isolation and functional characterization of monoclonal antibodies (mAbs) from two patients infected with CHIKV in the Dominican Republic. Single B cell sorting yielded a panel of 46 human mAbs of diverse germline lineages that targeted epitopes within the E1 or E2 glycoproteins. MAbs that recognized either E1 or E2 proteins exhibited neutralizing activity. Viral escape mutations localized the binding epitopes for two E1 mAbs to sites within domain I or the linker between domains I and III; and for two E2 mAbs between the ß-connector region and the B-domain. Two of the E2-specific mAbs conferred protection in vivo in a stringent lethal challenge mouse model of CHIKV infection, whereas the E1 mAbs did not. These results provide insight into human antibody response to CHIKV and identify candidate mAbs for therapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chikungunya Fever/immunology , Chikungunya virus/immunology , Epitopes/immunology , Glycoproteins/immunology , Viral Envelope Proteins/immunology , Adult , Animals , Antibodies, Neutralizing/immunology , Chikungunya Fever/virology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR
10.
J Virol ; 93(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31341048

ABSTRACT

The rotavirus polymerase VP1 mediates all stages of viral RNA synthesis within the confines of subviral particles and while associated with the core shell protein VP2. Transcription (positive-strand RNA [+RNA] synthesis) by VP1 occurs within double-layered particles (DLPs), while genome replication (double-stranded RNA [dsRNA] synthesis) by VP1 occurs within assembly intermediates. VP2 is critical for VP1 enzymatic activity; yet, the mechanism by which the core shell protein triggers polymerase function remains poorly understood. Structural analyses of transcriptionally competent DLPs show that VP1 is located beneath the VP2 core shell and sits slightly off-center from each of the icosahedral 5-fold axes. In this position, the polymerase is contacted by the core shell at 5 distinct surface-exposed sites, comprising VP1 residues 264 to 267, 547 to 550, 614 to 620, 968 to 980, and 1022 to 1025. Here, we sought to test the functional significance of these VP2 contact sites on VP1 with regard to polymerase activity. We engineered 19 recombinant VP1 (rVP1) proteins that contained single- or multipoint alanine mutations within each individual contact site and assayed them for the capacity to synthesize dsRNA in vitro in the presence of rVP2. Three rVP1 mutants (E265A/L267A, R614A, and D971A/S978A/I980A) exhibited diminished in vitro dsRNA synthesis. Despite their loss-of-function phenotypes, the mutants did not show major structural changes in silico, and they maintained their overall capacity to bind rVP2 in vitro via their nonmutated contact sites. These results move us toward a mechanistic understanding of rotavirus replication and identify precise VP2-binding sites on the polymerase surface that are critical for its enzymatic activation.IMPORTANCE Rotaviruses are important pathogens that cause severe gastroenteritis in the young of many animals. The viral polymerase VP1 mediates all stages of viral RNA synthesis, and it requires the core shell protein VP2 for its enzymatic activity. Yet, there are several gaps in knowledge about how VP2 engages and activates VP1. Here, we probed the functional significance of 5 distinct VP2 contact sites on VP1 that were revealed through previous structural studies. Specifically, we engineered alanine amino acid substitutions within each of the 5 VP1 regions and assayed the mutant polymerases for the capacity to synthesize RNA in the presence of VP2 in a test tube. Our results identified residues within 3 of the VP2 contact sites that are critical for robust polymerase activity. These results are important because they enhance the understanding of a key step of the rotavirus replication cycle.


Subject(s)
Capsid Proteins/genetics , Mutant Proteins/chemistry , RNA, Double-Stranded/chemistry , RNA, Viral/chemistry , RNA-Dependent RNA Polymerase/chemistry , Rotavirus , Amino Acid Sequence , Binding Sites , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Loss of Function Mutation , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Conformation , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Rotavirus/genetics , Rotavirus/metabolism
11.
J Interferon Cytokine Res ; 36(1): 62-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26430781

ABSTRACT

The omental fat band (OFB) is the predominant site for metastatic seeding of ovarian cancer. Previously, we highlighted the influx and accumulation of neutrophils and macrophages in the OFB following syngeneic ovarian cancer cell seeding as an important factor in the development of a protumorigenic cascade. Here we investigated localized immunomodulation as a means of promoting a successful protective response. As an important TH1-type immunomodulator, interleukin (IL)-12 has previously been investigated clinically as an anticancer therapeutic. However, systemic IL-12 administration was associated with serious side effects, galvanizing the development of immune or accessory cells engineered to express secreted or membrane-bound IL-12 (mbIL-12). Using an mbIL-12-expressing cell variant, we demonstrate that localized IL-12 in the tumor microenvironment significantly delays disease development. The mbIL-12-mediated decrease in tumor burden was associated with a significant reduction in neutrophil and macrophage infiltration in the OFB, and correlated with a reduced expression of neutrophil and macrophage chemoattractants (CXCL1, -2, -3 and CCL2, -7). Vaccination with mitotically impaired tumor cells did not confer protection against subsequent tumor challenge, indicating that IL-12 did not impact the immunogenicity of the cancer cells. Our findings are in agreement with previous reports suggesting that IL-12 may hold promise when delivered in a targeted and sustained manner to the omental microenvironment. Furthermore, resident cells within the omental microenvironment may provide a reservoir that can be activated and mobilized to prevent metastatic seeding within the peritoneum and, therefore, may be targets for chemotherapeutics.


Subject(s)
Carcinogenesis/immunology , Immunomodulation/genetics , Interleukin-12/immunology , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Female , Gene Expression Regulation , Humans , Interleukin-12/genetics , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/pathology , Omentum/immunology , Omentum/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/immunology , Ovary/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , Signal Transduction , Tumor Microenvironment/genetics
12.
Cancer Prev Res (Phila) ; 6(11): 1182-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24022590

ABSTRACT

Ovarian cancer is an insidious and aggressive disease of older women, typically undiscovered before peritoneal metastasis due to its asymptomatic nature and lack of early detection tools. Epidemiologic studies suggest that child-bearing (parity) is associated with decreased ovarian cancer risk, although the molecular mechanisms responsible for this phenomenon have not been delineated. Ovarian cancer preferentially metastasizes to the omental fat band (OFB), a secondary lymphoid organ that aids in filtration of the peritoneal serous fluid (PSF) and helps combat peritoneal infections. In the present study, we assessed how parity and age impact the immune compositional profile in the OFB of mice, both in the homeostatic state and as a consequence of peritoneal implantation of ovarian cancer. Using fluorescence-activated cell sorting analysis and quantitative real-time PCR, we found that parity was associated with a significant reduction in omental monocytic subsets and B1-B lymphocytes, correlating with reduced homeostatic expression levels of key chemoattractants and polarization factors (Ccl1, Ccl2, Arg1, and Cxcl13). Of note, parous animals exhibited significantly reduced tumor burden following intraperitoneal implantation compared with nulliparous animals. This was associated with a reduction in tumor-associated neutrophils and macrophages, as well as in the expression levels of their chemoattractants (Cxcl1 and Cxcl5) in the OFB and PSF. These findings define a preexisting "parity-associated microenvironmental niche" in the OFB that is refractory to metastatic tumor seeding and outgrowth. Future studies designed to manipulate this niche may provide a novel means to mitigate peritoneal dissemination of ovarian cancer.


Subject(s)
Omentum/pathology , Ovarian Neoplasms/pathology , Ovary/pathology , Parity/physiology , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Tumor Microenvironment , Animals , Biomarkers/metabolism , Cell Communication , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Omentum/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Peritoneal Neoplasms/metabolism , Pregnancy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
PLoS One ; 8(6): e66477, 2013.
Article in English | MEDLINE | ID: mdl-23776677

ABSTRACT

White adipose tissue (WAT) is a multi-faceted endocrine organ involved in energy storage, metabolism, immune function and disease pathogenesis. In contrast to subcutaneous fat, visceral fat (V-WAT) has been associated with numerous diseases and metabolic disorders, indicating specific functions related to anatomical location. Although visceral depots are often used interchangeably in V-WAT-associated disease studies, there has been a recent subdivision of V-WAT into "true visceral" and non-visceral intra-abdominal compartments. These were associated with distinct physiological roles, illustrating a need for depot-specific information. Here, we use FACS analysis to comparatively characterize the leukocyte and progenitor populations in the stromal vascular fraction (SVF) of peritoneal serous fluid (PSF), parametrial (pmWAT), retroperitoneal (rpWAT), and omental (omWAT) adipose tissue from seven-month old C57BL/6 female mice. We found significant differences in SVF composition between all four microenvironments. PSF SVF was comprised almost entirely of CD45(+) leukocytes (>99%), while omWAT contained less, but still almost two-fold more leukocytes than pmWAT and rpWAT (75%, 38% and 38% respectively; p<0.01). PmWAT was composed primarily of macrophages, whereas rpWAT more closely resembled omWAT, denoted by high levels of B1 B-cell and monocyte populations. Further, omWAT harbored significantly higher proportions of T-cells than the other tissues, consistent with its role as a secondary lymphoid organ. These SVF changes were also reflected in the gene expression profiles of the respective tissues. Thus, intra-abdominal fat pads represent independent immunomodulatory microenvironments and should be evaluated as distinct entities with unique contributions to physiological and pathological processes.


Subject(s)
Cellular Microenvironment/immunology , Immunomodulation/immunology , Intra-Abdominal Fat/immunology , Models, Animal , Adipose Tissue/cytology , Analysis of Variance , Animals , Ascitic Fluid/chemistry , Ascitic Fluid/cytology , Female , Flow Cytometry , Gene Expression Profiling , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
14.
Ann N Y Acad Sci ; 1122: 219-30, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18077575

ABSTRACT

In engineering and materials science, nanotechnology has provided many advances that effectively reduce oxidative damage generated by free radical production. Despite such advances, there has been little application to biomedical problems. Increased oxidative stress and free radical production are associated with neurodegenerative conditions, including aging, trauma, Alzheimer's and Parkinson's diseases, and many others. The antioxidant properties of cerium oxide nanoparticles show promise in the treatment of such diseases. Here, we summarize the work on the biological antioxidant actions of cerium oxide nanoparticles in extension of cell and organism longevity, protection against free radical insult, and protection against trauma-induced neuronal damage. We discuss establishment of effective dosing parameters, along with the physicochemical properties that regulate the pharmacological action of these new nanomaterials. Taken together, these studies suggest that nanotechnology can take pharmacological treatment to a new level, with a novel generation of nanopharmaceuticals.


Subject(s)
Cerium/therapeutic use , Nanomedicine/methods , Neurodegenerative Diseases/therapy , Neuroprotective Agents/therapeutic use , Animals , Free Radicals/toxicity , Humans
15.
Nanomedicine (Lond) ; 1(4): 399-412, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17716143

ABSTRACT

Nanotechnology has made significant advances in the reduction of free radical damage in the field of materials science. Cross-disciplinary interactions and the application of this technology to biological systems has led to the elucidation of novel nanoparticle antioxidants, which are the subject of this review. Recent reports suggest that cerium oxide and other nanoparticles are potent, and probably regenerative, free radical scavengers in vitro and in vivo. The neuroprotective, longevity-enhancing and anti-inflammatory properties of nanoparticles are summarized and hypotheses regarding their unique mechanism of action are presented. The chemical and physical properties of antioxidant nanoparticles are discussed in an interdisciplinary manner, with emphasis on biological properties and biomedical applications. Additionally, the need for alterations in traditional pharmacological parameters of dose and absorption, distribution, metabolism, and excretion are discussed and future directions necessary for bringing nanoparticle antioxidants into the realm of clinical reality are presented.


Subject(s)
Antioxidants/chemistry , Antioxidants/therapeutic use , Drug Design , Nanomedicine/methods , Nanomedicine/trends , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...