Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Aging Cell ; 22(8): e13897, 2023 08.
Article in English | MEDLINE | ID: mdl-37272263

ABSTRACT

Developing accurate methods to quantify age-related muscle loss (sarcopenia) could greatly accelerate development of therapies to treat muscle loss in the elderly, as current methods are inaccurate or expensive. The current gold standard method for quantifying sarcopenia is dual-energy X-ray absorptiometry (DXA) but does not measure muscle directly-it is a composite measure quantifying "lean mass" (muscle) excluding fat and bone. In humans, DXA overestimates muscle mass, which has led to erroneous conclusions about the importance of skeletal muscle in human health and disease. In animal models, DXA is a popular method for measuring lean mass. However, instrumentation is expensive and is potentially limited by anesthesia concerns. Recently, the D3 -creatine (D3 Cr) dilution method for quantifying muscle mass was developed in humans and rats. This method is faster, cheaper, and more accurate than DXA. Here, we demonstrate that the D3 Cr method is a specific assay for muscle mass in mice, and we test associations with DXA and body weight. We evaluated the D3 Cr method compared to DXA-determined lean body mass (LBM) in aged mice and reported that DXA consistently overestimates muscle mass with age. Overall, we provide evidence that the D3 Cr dilution method directly measures muscle mass in mice. Combined with its ease of use, accessibility, and non-invasive nature, the method may prove to more quickly advance development of preclinical therapies targeting sarcopenia.


Subject(s)
Body Composition , Body Weights and Measures , Creatinine , Muscle, Skeletal , Absorptiometry, Photon , Animals , Mice , Male , Female , Mice, Inbred C57BL , Creatinine/urine , Body Weights and Measures/methods
2.
Cell Rep ; 31(2): 107494, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294445

ABSTRACT

Paradoxically, early host responses to infection include the upregulation of the antiphagocytic molecule, CD47. This suggests that CD47 blockade could enhance antigen presentation and subsequent immune responses. Indeed, mice treated with anti-CD47 monoclonal antibody following lymphocytic choriomeningitis virus infections show increased activation of both macrophages and dendritic cells (DCs), enhancement of the kinetics and potency of CD8+ T cell responses, and significantly improved virus control. Treatment efficacy is critically dependent on both APCs and CD8+ T cells. In preliminary results from one of two cohorts of humanized mice infected with HIV-1 for 6 weeks, CD47 blockade reduces plasma p24 levels and restores CD4+ T cell counts. The results indicate that CD47 blockade not only enhances the function of innate immune cells but also links to adaptive immune responses through improved APC function. As such, immunotherapy by CD47 blockade may have broad applicability to treat a wide range of infectious diseases.


Subject(s)
CD47 Antigen/immunology , CD47 Antigen/metabolism , Virus Diseases/immunology , Adaptive Immunity/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Female , HEK293 Cells , Humans , Immunity, Innate/immunology , Immunotherapy/methods , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
3.
Viruses ; 11(3)2019 03 13.
Article in English | MEDLINE | ID: mdl-30871222

ABSTRACT

Although antiretroviral therapy (ART) greatly suppresses HIV replication, lymphoid tissues remain a sanctuary site where the virus may replicate. Tracking the earliest steps of HIV spread from these cellular reservoirs after drug cessation is pivotal for elucidating how infection can be prevented. In this study, we developed an in vivo model of HIV persistence in which viral replication in the lymphoid compartments of humanized mice was inhibited by the HIV reverse transcriptase inhibitor 4'-ethynyl-2-fluoro-2'-deoxyadenosine (EFdA) to very low levels, which recapitulated ART-suppression in HIV-infected individuals. Using a combination of RNAscope in situ hybridization (ISH) and immunohistochemistry (IHC), we quantitatively investigated the distribution of HIV in the lymphoid tissues of humanized mice during active infection, EFdA suppression, and after drug cessation. The lymphoid compartments of EFdA-suppressed humanized mice harbored very rare transcription/translation-competent HIV reservoirs that enable viral rebound. Our data provided the visualization and direct measurement of the early steps of HIV reservoir expansion within anatomically intact lymphoid tissues soon after EFdA cessation and suggest a strategy to enhance therapeutic approaches aimed at eliminating the HIV reservoir.


Subject(s)
Deoxyadenosines/pharmacology , Disease Reservoirs/virology , HIV-1/physiology , Lymphoid Tissue/virology , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication/drug effects , Animals , Disease Models, Animal , Female , HIV Infections/drug therapy , HIV-1/drug effects , Humans , Mice , Mice, Transgenic
4.
Antimicrob Agents Chemother ; 59(7): 4190-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941222

ABSTRACT

Like normal cellular nucleosides, the nucleoside reverse transcriptase (RT) inhibitor (NRTI) 4'-ethynyl-2-fluoro-2'-deoxyadenosine (EFdA) has a 3'-hydroxyl moiety, and yet EFdA is a highly potent inhibitor of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication with activity against a broad range of clinically important drug-resistant HIV isolates. We evaluated the anti-HIV activity of EFdA in primary human cells and in HIV-infected humanized mice. EFdA exhibited excellent potency against HIVJR-CSF in phytohemagglutinin-stimulated peripheral blood mononuclear cells (PBMCs), with a 50% inhibitory concentration of 0.25 nM and a selectivity index of 184,000; similar antiviral potency was found against 12 different HIV clinical isolates from multiple clades (A, B, C, D, and CRF01_AE). EFdA was readily absorbed after oral dosing (5 mg/kg of body weight) in both mice and the rhesus macaque, with micromolar levels of the maximum concentration of drug in serum (Cmax) attained at 30 min and 90 min, respectively. Trough levels were at or above 90% inhibitory concentration (IC90) levels in the macaque at 24 h, suggesting once-daily dosing. EFdA showed reasonable penetration of the blood-brain barrier in the rhesus macaque, with cerebrospinal fluid levels at approximately 25% of plasma levels 8 h after single oral dosing. Rhesus PBMCs isolated 24 h following a single oral dose of 5 mg/kg EFdA were refractory to SIV infection due to sufficiently high intracellular EFdA-triphosphate levels. The intracellular half-life of EFdA-triphosphate in PBMCs was determined to be >72 h following a single exposure to EFdA. Daily oral administration of EFdA at low dosage levels (1 to 10 mg/kg/day) was highly effective in protecting humanized mice from HIV infection, and 10 mg/kg/day oral EFdA completely suppressed HIV RNA to undetectable levels within 2 weeks of treatment.


Subject(s)
Anti-HIV Agents/therapeutic use , Diazonium Compounds/therapeutic use , Farnesol/analogs & derivatives , HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/therapeutic use , Animals , Anti-HIV Agents/administration & dosage , Blood-Brain Barrier , Diazonium Compounds/administration & dosage , Diazonium Compounds/pharmacokinetics , Farnesol/administration & dosage , Farnesol/pharmacokinetics , Farnesol/therapeutic use , Flow Cytometry , HIV Infections/virology , HIV-1/drug effects , Half-Life , Humans , In Vitro Techniques , Macaca mulatta , Mice , Mice, SCID , Monocytes/drug effects , Monocytes/virology , RNA, Viral/biosynthesis , RNA, Viral/drug effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Simian Immunodeficiency Virus , Viremia/drug therapy , Viremia/virology
5.
Virology ; 462-463: 115-25, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24971704

ABSTRACT

Highly potent broadly neutralizing human monoclonal antibodies hold promise for HIV prophylaxis and treatment. We used the SCID-hu Thy/Liv and BLT humanized mouse models to study the efficacy of these antibodies, primarily PG16, against HIV-1 clades A, B, and C. PG16 targets a conserved epitope in the V1/V2 region of gp120 common to 70-80% of HIV-1 isolates from multiple clades and has extremely potent in vitro activity against HIVJR-CSF. PG16 was highly efficacious in SCID-hu mice as a single intraperitoneal administration the day before inoculation of R5-tropic HIV directly into their Thy/Liv implants and demonstrated even greater efficacy if PG16 administration was continued after Thy/Liv implant HIV inoculation. However, PG16 as monotherapy had no activity in humanized mice with established R5-tropic HIV infection. These results provide evidence of tissue penetration of the antibodies, which could aid in their ability to prevent infection if virus crosses the mucosal barrier.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , HIV Antibodies/therapeutic use , HIV Infections/therapy , HIV-1/immunology , Animals , Disease Models, Animal , HIV Envelope Protein gp120/immunology , Male , Mice , Mice, SCID , Treatment Outcome
6.
Antimicrob Agents Chemother ; 56(4): 2162-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22252805

ABSTRACT

PC-1505 is a C34 peptide derived from the heptad repeat 2 region of HIV-1 gp41 conjugated to human serum albumin for sustained in vivo activity. One single preexposure dose of PC-1505 reduced viral RNA in HIV-1-infected SCID-hu Thy/Liv mice by 3.3 log10 and protected T cells from virus-mediated depletion. In contrast, a single preexposure dose of Truvada reduced viral RNA by only 0.8 log10 and was substantially less effective in preventing T cell depletion.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV Infections/prevention & control , Peptides/pharmacology , Animals , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Genes, MHC Class I/genetics , HIV Core Protein p24/blood , HIV Fusion Inhibitors/chemistry , HIV Infections/virology , Mice , Mice, SCID , Organophosphorus Compounds/pharmacology , Peptides/chemistry , RNA, Viral/blood , Thymocytes/drug effects , Thymocytes/metabolism , Thymocytes/virology
7.
Virology ; 417(1): 154-60, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21684569

ABSTRACT

Humanized Bone marrow/Liver/Thymus (BLT) mice recapitulate the mucosal transmission of HIV, permitting study of early events in HIV pathogenesis and evaluation of preexposure prophylaxis methods to inhibit HIV transmission. Human hematopoiesis is reconstituted in NOD-scid mice by implantation of human fetal liver and thymus tissue to generate human T cells plus intravenous injection of autologous liver-derived CD34(+) hematopoietic stem cells to engraft the mouse bone marrow. In side-by-side comparisons, we show that NOD-scid mice homozygous for a deletion of the IL-2Rγ-chain (NOD-scid IL-2Rγ(-/-)) are far superior to NOD-scid mice in both their peripheral blood reconstitution with multiple classes of human leukocytes (e.g., a mean of 182 versus 14 CD4(+) T cells per µl 12 weeks after CD34(+) injection) and their susceptibility to intravaginal HIV exposure (84% versus 11% viremic mice at 4 weeks). These results should speed efforts to obtain preclinical animal efficacy data for new HIV drugs and microbicides.


Subject(s)
HIV Infections/transmission , HIV/immunology , Interleukin Receptor Common gamma Subunit/physiology , Leukocytes/physiology , Animals , Antigens, CD34 , Disease Susceptibility , Female , HIV/physiology , Humans , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Transplantation, Heterologous , Vagina
8.
Science ; 330(6011): 1695-9, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-21164017

ABSTRACT

Although the mammalian immune system is generally thought to develop in a linear fashion, findings in avian and murine species argue instead for the developmentally ordered appearance (or "layering") of distinct hematopoietic stem cells (HSCs) that give rise to distinct lymphocyte lineages at different stages of development. Here we provide evidence of an analogous layered immune system in humans. Our results suggest that fetal and adult T cells are distinct populations that arise from different populations of HSCs that are present at different stages of development. We also provide evidence that the fetal T cell lineage is biased toward immune tolerance. These observations offer a mechanistic explanation for the tolerogenic properties of the developing fetus and for variable degrees of immune responsiveness at birth.


Subject(s)
Cell Lineage , Fetus/immunology , Hematopoietic Stem Cells/physiology , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Adult , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Aging/immunology , Animals , Bone Marrow/embryology , Bone Marrow Cells , CD4-Positive T-Lymphocytes/immunology , Cytoprotection , Fetus/cytology , Gene Expression , Humans , Liver/cytology , Liver/embryology , Lymphocyte Activation , Lymphopoiesis , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/cytology , Thymus Gland/immunology , Young Adult
9.
J Biol Chem ; 283(49): 34045-52, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18809675

ABSTRACT

Entry inhibitors of human immunodeficiency virus, type 1 (HIV-1) have been the focus of much recent research. C34, a potent fusion inhibitor derived from the HR2 region of gp41, was engineered into a 1:1 human serum albumin conjugate through stable covalent attachment of a maleimido-C34 analog onto cysteine 34 of albumin. This bioconjugate, PC-1505, was designed to require less frequent dosing and less peptide than T-20 and was assessed for its antifusogenic activity both in vitro and in vivo in the SCID-hu Thy/Liv mouse model. PC-1505 was essentially equipotent to the original C34 peptide and to T-20 in vitro. In HIV-1-infected SCID-hu Thy/Liv mice, T-20 lost activity with infrequent dosing, whereas the antiviral potency of PC-1505 was sustained, and PC-1505 was active against T-20-resistant ("DIV") virus with a G36D substitution in gp41. The in vivo results are the direct result of a significantly improved pharmacokinetic profile for the C34 peptide following albumin conjugation. Contrary to previous reports that the gp41 NHR trimer is poorly accessible to C34 fused to protein cargoes of increasing size (Hamburger, A. E., Kim, S., Welch, B. D., and Kay, M. S. (2005) J. Biol. Chem. 280, 12567-12572), these results are the first demonstration of the capacity for a large, endogenous serum protein to gain unobstructed access to the transient gp41 intermediates that exist during the HIV fusion process, and it supports further development of albumin conjugation as a promising approach to inhibit HIV-1 entry.


Subject(s)
Albumins/chemistry , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV-1/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/pharmacology , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Disease Models, Animal , Enfuvirtide , HIV Fusion Inhibitors/chemistry , Mice , Mice, SCID , Molecular Sequence Data , Peptides/chemistry , Rats , Rats, Sprague-Dawley
10.
PLoS One ; 2(7): e655, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17668043

ABSTRACT

BACKGROUND: The SCID-hu Thy/Liv mouse model of HIV-1 infection is a useful platform for the preclinical evaluation of antiviral efficacy in vivo. We performed this study to validate the model with representatives of all four classes of licensed antiretrovirals. METHODOLOGY/PRINCIPAL FINDINGS: Endpoint analyses for quantification of Thy/Liv implant viral load included ELISA for cell-associated p24, branched DNA assay for HIV-1 RNA, and detection of infected thymocytes by intracellular staining for Gag-p24. Antiviral protection from HIV-1-mediated thymocyte depletion was assessed by multicolor flow cytometric analysis of thymocyte subpopulations based on surface expression of CD3, CD4, and CD8. These mice can be productively infected with molecular clones of HIV-1 (e.g., the X4 clone NL4-3) as well as with primary R5 and R5X4 isolates. To determine whether results in this model are concordant with those found in humans, we performed direct comparisons of two drugs in the same class, each of which has known potency and dosing levels in humans. Here we show that second-generation antiretrovirals were, as expected, more potent than their first-generation predecessors: emtricitabine was more potent than lamivudine, efavirenz was more potent than nevirapine, and atazanavir was more potent than indinavir. After interspecies pharmacodynamic scaling, the dose ranges found to inhibit viral replication in the SCID-hu Thy/Liv mouse were similar to those used in humans. Moreover, HIV-1 replication in these mice was genetically stable; treatment of the mice with lamivudine did not result in the M184V substitution in reverse transcriptase, and the multidrug-resistant NY index case HIV-1 retained its drug-resistance substitutions. CONCLUSION: Given the fidelity of such comparisons, we conclude that this highly reproducible mouse model is likely to predict clinical antiviral efficacy in humans.


Subject(s)
Anti-HIV Agents/therapeutic use , Mice, SCID/genetics , Severe Combined Immunodeficiency/genetics , Acquired Immunodeficiency Syndrome/genetics , Animals , Anti-Retroviral Agents/therapeutic use , DNA, Viral/genetics , Disease Models, Animal , HIV Infections/drug therapy , HIV Infections/genetics , HIV-1/drug effects , HIV-1/genetics , HIV-1/physiology , Histocompatibility Antigens Class I/immunology , Humans , Mice , RNA, Viral/genetics , Severe Combined Immunodeficiency/immunology , Viral Load/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...