Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Arthroscopy ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38942098

ABSTRACT

PURPOSE: The aim of this study was to present the short-term outcomes of arthroscopic in-situ biceps tenodesis combined with partial rotator cuff repair in patients with massive irreparable rotator cuff tears (MIRCT) and minimal arthritis. METHODS: A retrospective review was conducted utilizing prospectively maintained institutional databases to identify patients who had undergone a partial rotator cuff repair with in-situ biceps tenodesis between March 2017 to December 2022. Patients were included if they (1) were diagnosed pre- or intra-operatively with MIRCT, and (2) had complete pre-operative and minimum 1-year post-operative patient-reported outcome measures. RESULTS: Thirty-nine patients met the eligibility criteria and were included for analysis. The mean age of the study participants was 65 ± 7 years (range: 46 - 76) with 76.9% (n = 30) being male. The average follow-up was 21 ± 12 months (range: 12 - 58). Patients experienced significant improvement in visual analog scale for pain (VAS), American Shoulder and Elbow Surgeons (ASES) score, and subjective assessment numeric evaluation (SANE) score (p < 0.001 for all comparisons). The pre- to post-operative improvement was 3.1 ± 2.3 for VAS, 27.5 ± 20.6 for ASES, and 31.3 ± 24.8 for SANE. Postoperatively, the average scores for VAS, ASES, and SANE were 1.3 ± 1.5, 79.5 ± 17.0, and 69.6 ± 20.1, respectively. Twenty-six patients (66.7%) achieved the minimal clinically important difference (MCID) for VAS, thirty-three patients (84.6%) achieved the MCID for ASES, and thirty patients (76.9%) achieved the MCID for SANE.. CONCLUSIONS: Partial rotator cuff repair with in-situ biceps tenodesis is an effective treatment for MIRCT, leading to significant improvements in patient-reported outcome and range of motion measures compared to preoperative conditions.

2.
Arthrosc Tech ; 11(11): e1951-e1956, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36457383

ABSTRACT

This article describes a simple all soft tissue technique for arthroscopic long head of the biceps tenodesis to the subscapularis tendon using posterior and anterior portals. The technique uses a PDS suture that assists in passing a braided suture through both the biceps tendon and the subscapularis to allow for the desired tenodesis. This technique is simple, safe, efficient, and less costly than other techniques.

3.
Front Immunol ; 13: 840976, 2022.
Article in English | MEDLINE | ID: mdl-35572573

ABSTRACT

A better understanding of the impact of early innate immune responses after vaccine priming on vaccine-elicited adaptive immune responses could inform rational design for effective HIV vaccines. The current study compared the whole blood molecular immune signatures of a 3M-052-SE adjuvanted HIV Env protein vaccine to a regimen combining the adjuvanted Env protein with simultaneous administration of a modified Vaccinia Ankara vector expressing HIV Env in infant rhesus macaques at days 0, 1, and 3 post vaccine prime. Both vaccines induced a rapid innate response, evident by elevated inflammatory plasma cytokines and altered gene expression. We identified 25 differentially-expressed genes (DEG) on day 1 compared to day 0 in the HIV protein vaccine group. In contrast, in the group that received both the Env protein and the MVA-Env vaccine only two DEG were identified, implying that the MVA-Env modified the innate response to the adjuvanted protein vaccine. By day 3, only three DEG maintained altered expression, indicative of the transient nature of the innate response. The DEG represented immune pathways associated with complement activation, type I interferon and interleukin signaling, pathogen sensing, and induction of adaptive immunity. DEG expression on day 1 was correlated to Env-specific antibody responses, in particular antibody-dependent cytotoxicity responses at week 34, and Env-specific follicular T helper cells. Results from network analysis supported the interaction of DEG and their proteins in B cell activation. These results emphasize that vaccine-induced HIV-specific antibody responses can be optimized through the modulation of the innate response to the vaccine prime.


Subject(s)
AIDS Vaccines , HIV Antibodies/blood , HIV Infections , Viral Envelope Proteins/immunology , Adjuvants, Immunologic/pharmacology , Animals , Gene Products, env , HIV Antibodies/immunology , HIV Infections/prevention & control , Humans , Macaca mulatta , Vaccination , Vaccinia virus/genetics
4.
mSphere ; 7(1): e0083921, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35196125

ABSTRACT

Improved access to antiretroviral therapy (ART) and antenatal care has significantly reduced in utero and peripartum mother-to-child human immunodeficiency virus (HIV) transmission. However, as breast milk transmission of HIV still occurs at an unacceptable rate, there remains a need to develop an effective vaccine for the pediatric population. Previously, we compared different HIV vaccine strategies, intervals, and adjuvants in infant rhesus macaques to optimize the induction of HIV envelope (Env)-specific antibodies with Fc-mediated effector function. In this study, we tested the efficacy of an optimized vaccine regimen against oral simian-human immunodeficiency virus (SHIV) acquisition in infant macaques. Twelve animals were immunized with 1086.c gp120 protein adjuvanted with 3M-052 in stable emulsion and modified vaccinia Ankara (MVA) virus expressing 1086.c HIV Env. Twelve control animals were immunized with empty MVA. The vaccine prime was given within 10 days of birth, with booster doses being administered at weeks 6 and 12. The vaccine regimen induced Env-specific plasma IgG antibodies capable of antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Beginning at week 15, infants were exposed orally to escalating doses of heterologous SHIV-1157(QNE)Y173H once a week until infected. Despite the induction of strong Fc-mediated antibody responses, the vaccine regimen did not reduce the risk of infection or time to acquisition compared to controls. However, among vaccinated animals, ADCC postvaccination and postinfection was associated with reduced peak viremia. Thus, nonneutralizing Env-specific antibodies with Fc effector function elicited by this vaccine regimen were insufficient for protection against heterologous oral SHIV infection shortly after the final immunization but may have contributed to control of viremia. IMPORTANCE Women of childbearing age are three times more likely to contract HIV infection than their male counterparts. Poor HIV testing rates coupled with low adherence to antiretroviral therapy (ART) result in a high risk of mother-to-infant HIV transmission, especially during the breastfeeding period. A preventative vaccine could curb pediatric HIV infections, reduce potential health sequalae, and prevent the need for lifelong ART in this population. The results of the current study imply that the HIV Env-specific IgG antibodies elicited by this candidate vaccine regimen, despite a high magnitude of Fc-mediated effector function but a lack of neutralizing antibodies and polyfunctional T cell responses, were insufficient to protect infant rhesus macaques against oral virus acquisition.


Subject(s)
AIDS Vaccines , HIV Infections , HIV-1 , Simian Immunodeficiency Virus , Animals , Child , Female , HIV Antibodies , Humans , Immunoglobulin G , Infectious Disease Transmission, Vertical/prevention & control , Macaca mulatta , Male , Pregnancy , Vaccinia virus , Viremia
5.
PLoS One ; 16(12): e0256885, 2021.
Article in English | MEDLINE | ID: mdl-34972105

ABSTRACT

Different HIV vaccine regimens elicit distinct plasma antibody responses in both human and nonhuman primate models. Previous studies in human and non-human primate infants showed that adjuvants influenced the quality of plasma antibody responses induced by pediatric HIV envelope vaccine regimens. We recently reported that use of the 3M052-SE adjuvant and longer intervals between vaccinations are associated with higher magnitude of antibody responses in infant rhesus macaques. However, the impact of different adjuvants in HIV vaccine regimens on the developing infant B cell receptor (BCR) repertoire has not been studied. This study evaluated whether pediatric HIV envelope vaccine regimens with different adjuvants induced distinct antigen-specific memory B cell repertoires and whether specific immunoglobulin (Ig) immunogenetic characteristics are associated with higher magnitude of plasma antibody responses in vaccinated infant rhesus macaques. We utilized archived preclinical pediatric HIV vaccine studies PBMCs and tissue samples from 19 infant rhesus macaques immunized either with (i) HIV Env protein with a squalene adjuvant, (ii) MVA-HIV and Env protein co-administered using a 3-week interval, (iii) MVA-HIV prime/ protein boost with an extended 6-week interval between immunizations, or (iv) with HIV Env administered with 3M-052-SE adjuvant. Frequencies of vaccine-elicited HIV Env-specific memory B cells from PBMCs and tissues were similar across vaccination groups (frequency range of 0.06-1.72%). There was no association between vaccine-elicited antigen-specific memory B cell frequencies and plasma antibody titer or avidity. Moreover, the epitope specificity and Ig immunogenetic features of vaccine-elicited monoclonal antibodies did not differ between the different vaccine regimens. These data suggest that pediatric HIV envelope vaccine candidates with different adjuvants that previously induced higher magnitude and quality of plasma antibody responses in infant rhesus macaques were not driven by distinct antigen-specific memory BCR repertoires.


Subject(s)
AIDS Vaccines/blood , AIDS Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Antibody Formation/immunology , Receptors, Antigen, B-Cell/metabolism , env Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibody Formation/drug effects , Child , Complementarity Determining Regions , Epitopes/immunology , Humans , Immunization , Immunoglobulin Heavy Chains/metabolism , Immunologic Memory/drug effects , Macaca mulatta , Somatic Hypermutation, Immunoglobulin , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
6.
Sci Immunol ; 6(60)2021 06 15.
Article in English | MEDLINE | ID: mdl-34131024

ABSTRACT

The inclusion of infants in the SARS-CoV-2 vaccine roll-out is important to prevent severe complications of pediatric SARS-CoV-2 infections and to limit transmission and could possibly be implemented via the global pediatric vaccine schedule. However, age-dependent differences in immune function require careful evaluation of novel vaccines in the pediatric population. Toward this goal, we assessed the safety and immunogenicity of two SARS-CoV-2 vaccines. Two groups of 8 infant rhesus macaques (RMs) were immunized intramuscularly at weeks 0 and 4 with stabilized prefusion SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or the purified S protein mixed with 3M-052, a synthetic TLR7/8 agonist in a squalene emulsion (Protein+3M-052-SE). Neither vaccine induced adverse effects. Both vaccines elicited high magnitude IgG binding to RBD, N terminus domain, S1, and S2, ACE2 blocking activity, and high neutralizing antibody titers, all peaking at week 6. S-specific memory B cells were detected by week 4 and S-specific T cell responses were dominated by the production of IL-17, IFN-γ, or TNF-α. Antibody and cellular responses were stable through week 22. The immune responses for the mRNA-LNP vaccine were of a similar magnitude to those elicited by the Moderna mRNA-1273 vaccine in adults. The S-2P mRNA-LNP and Protein-3M-052-SE vaccines were well-tolerated and highly immunogenic in infant RMs, providing proof-of concept for a pediatric SARS-CoV-2 vaccine with the potential for durable immunity that might decrease the transmission of SARS-CoV-2 and mitigate the ongoing health and socioeconomic impacts of COVID-19.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/prevention & control , Macaca mulatta , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/immunology
7.
bioRxiv ; 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33851156

ABSTRACT

Early life SARS-CoV-2 vaccination has the potential to provide lifelong protection and achieve herd immunity. To evaluate SARS-CoV-2 infant vaccination, we immunized two groups of 8 infant rhesus macaques (RMs) at weeks 0 and 4 with stabilized prefusion SARS-CoV-2 S-2P spike (S) protein, either encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or mixed with 3M-052-SE, a TLR7/8 agonist in a squalene emulsion (Protein+3M-052-SE). Neither vaccine induced adverse effects. High magnitude S-binding IgG and neutralizing infectious dose 50 (ID 50 ) >10 3 were elicited by both vaccines. S-specific T cell responses were dominated by IL-17, IFN- γ , or TNF- α . Antibody and cellular responses were stable through week 22. The S-2P mRNA-LNP and Protein-3M-052-SE vaccines are promising pediatric SARS-CoV-2 vaccine candidates to achieve durable protective immunity. ONE-SENTENCE SUMMARY: SARS-CoV-2 vaccines are well-tolerated and highly immunogenic in infant rhesus macaques.

8.
Curr Rev Musculoskelet Med ; 13(6): 748-756, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32827302

ABSTRACT

PURPOSE OF REVIEW: This article aims to provide a comprehensive understanding of the evaluation, diagnosis, and management of scapular dyskinesis and its impact on the kinetic chain in tennis athletes. RECENT FINDINGS: Optimal glenohumeral biomechanics are intimately associated with proper scapular motion and function. The tennis serve requires the scapula to act as a force transducer in the kinetic chain to convert potential energy generated in the lower extremities to kinetic energy in the upper extremity. Any aberration within this complex kinetic chain will result in force uncoupling and increases the potential for injury through compensatory mechanisms. Specifically, scapular dyskinesis has been associated with an increased risk of shoulder pain of up to 43% in overhead athletes. These pathologies include rotator cuff disease, subacromial and posterior impingement, labral injuries, and SLAP tears. Although the direct causality of these injuries remains controversial, multiple kinematic studies have demonstrated altered scapular positioning increasing the predilection for soft tissue pathology. The diagnosis of scapular dyskinesis is predicated upon a thorough history, physical examination, and observational analysis of key nodes in the kinetic chain during tennis activity. Conservative management remains the mainstay of treatment and consists of a graduated physical therapy regimen. Although shoulder pain in the overhead athletes is often multifactorial, early recognition and treatment of scapular dyskinesis generally carry a favorable prognosis and result in improved patient outcomes.

9.
mSphere ; 5(2)2020 03 25.
Article in English | MEDLINE | ID: mdl-32213623

ABSTRACT

The HIV epidemics in infants and adolescent women are linked. Young women of childbearing age are at high risk for HIV infection and, due to poor HIV testing rates and low adherence to antiretroviral therapy, are at high risk for mother-to-infant transmission. We hypothesize that HIV vaccine regimens initiated in early life would provide the necessary time frame to induce mature and highly functional Env-specific antibody responses that could potentially also protect against HIV acquisition later in life. The present study was designed to test two vaccine regimens, a clade C HIV Env protein vaccine (Env only) alone or combined with a modified vaccinia Ankara (MVA) vector expressing HIV Env (MVA/Env) for the induction and persistence of Env-specific antibody responses in an infant nonhuman primate model. Vaccination was initiated within the first week of life, with booster immunizations at weeks 6, 12, and 32. We demonstrate that both vaccine strategies were able to elicit durable Env-specific antibody responses that were enhanced by a late boost in infancy. Furthermore, we confirmed earlier data that intramuscular administration of the Env protein with the Toll-like receptor 7/8 (TLR7/8)-based adjuvant 3M-052 in stable emulsion (3M-052-SE) induced higher Env-specific antibody responses than vaccination with Env adjuvanted in Span85-Tween 80-squalene (STS) tested in a previous study. These results support the concept of early vaccination as a means to induce durable immune responses that may prevent HIV infection in adolescence at the onset of sexual debut.IMPORTANCE The majority of new HIV-1 infections occur in young adults, with adolescent women being 3 times more likely to acquire HIV than young men. Implementation of HIV prevention strategies has been less successful in this age group; thus, a vaccine given prior to adolescence remains a high priority. We propose that instead of starting HIV vaccination during adolescence, an HIV vaccine regimen initiated in early infancy, aligned with the well-accepted pediatric vaccine schedule and followed with booster immunizations, will provide an alternative means to reduce HIV acquisition in adolescence. Importantly, the long window of time between the first infant vaccine dose and the adolescence vaccine dose will allow for the maturation of highly functional HIV Env-specific antibody responses. Our study provides evidence that early life vaccination induces durable Env-specific plasma IgG responses that can be boosted to further improve the quality of the antibody response.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , Immunization, Secondary , Immunoglobulin G/immunology , AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Female , HIV Antibodies/blood , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1 , Immunization Schedule , Immunoglobulin G/blood , Macaca mulatta/immunology , Male , Vaccination
10.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31801861

ABSTRACT

Mother-to-child transmission of human immunodeficiency virus type 1 (HIV-1) via breastfeeding is responsible for nearly half of new infections of children with HIV. Although innate lymphoid cells (ILC) and natural killer (NK) cells are found throughout the oral mucosae, the effects of HIV/simian-human immunodeficiency virus (SHIV) in these tissues are largely unknown. To better understand the mechanics of postnatal transmission, we performed a comprehensive study of simian immunodeficiency virus (SIV)/SHIV-infected infant rhesus macaques (RM) and tracked changes in frequency, trafficking, and function of group 3 ILC (ILC3) and NK cells using polychromatic flow cytometry and cell stimulation assays in colon, tonsil, and oral lymph node samples. Infection led to a 3-fold depletion of ILC3 in the colon and an increase in the levels of NK cells in tonsils and oral lymph nodes. ILC3 and NK cells exhibited alterations in their trafficking repertoires as a result of infection, with increased expression of CD103 in colon NK cells and curtailment of CXCR3, and a significant decrease in α4ß7 expression in colon ILC3. SPICE analyses revealed that ILC3 and NK cells displayed distinct functional profiles by tissue in naive samples. Infection perturbed these profiles, with a nearly total loss of interleukin-22 (IL-22) production in the tonsil and colon; an increase in the levels of CD107a, gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α) from ILC3; and an increase in the levels of CD107a, macrophage inflammatory protein 1 beta (MIP-1ß), and TNF-α from NK cells. Collectively, these data reveal that lentivirus infection alters the frequencies, receptor repertoires, and functions of innate cells in the oral and gut mucosa of infants. Further study will be required to delineate the full extent of the effect that these changes have on oral and gut homeostasis, SHIV/SIV pathogenesis, and oral opportunistic disease.IMPORTANCE Vertical transmission of HIV from mother to child accounts for many of the new cases seen worldwide. There is currently no vaccine to mitigate this transmission, and there has been limited research on the effects that lentiviral infection has on the innate immune system in oral tissues of infected children. To fill this knowledge gap, our laboratory studied infant rhesus macaques to evaluate how acute SIV/SHIV infections impacted ILC3 and NK cells, which are immune cells critical for mucosal homeostasis and antimicrobial defense. Our data revealed that SIV/SHIV infection led to a depletion of ILC3 and an increase of NK cells and to a functional shift from a homeostatic to a multifunctional proinflammatory state. Taking the results together, we describe how lentiviral infection perturbs the oral and gastrointestinal mucosae of infant macaques through alterations of resident innate immune cells giving rise to chronic inflammation and potentially exacerbating morbidity and mortality in children living with HIV.


Subject(s)
Immunity, Innate , Killer Cells, Natural/immunology , Macaca mulatta/virology , Mucous Membrane/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Gastrointestinal Tract , HIV Infections/immunology , HIV-1/immunology , Infectious Disease Transmission, Vertical , Interferon-gamma/immunology , Interleukins/metabolism , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load , Interleukin-22
11.
mBio ; 10(5)2019 09 05.
Article in English | MEDLINE | ID: mdl-31488511

ABSTRACT

To achieve long-term viral remission in human immunodeficiency virus (HIV)-infected children, novel strategies beyond early antiretroviral therapy (ART) will be necessary. Identifying clinical predictors of the time to viral rebound upon ART interruption will streamline the development of novel therapeutic strategies and accelerate their evaluation in clinical trials. However, identification of these biomarkers is logistically challenging in infants, due to sampling limitations and the potential risks of treatment interruption. To facilitate the identification of biomarkers predicting viral rebound, we have developed an infant rhesus macaque (RM) model of oral simian-human immunodeficiency virus (SHIV) SHIV.CH505.375H.dCT challenge and analytical treatment interruption (ATI) after short-term ART. We used this model to characterize SHIV replication kinetics and virus-specific immune responses during short-term ART or after ATI and demonstrated plasma viral rebound in 5 out of 6 (83%) infants. We observed a decline in humoral immune responses and partial dampening of systemic immune activation upon initiation of ART in these infants. Furthermore, we monitored SHIV replication and rebound kinetics in infant and adult RMs and found that both infants and adults demonstrated equally potent virus-specific humoral immune responses. Finally, we validated our models by confirming a well-established correlate of the time to viral rebound, namely, the pre-ART plasma viral load, as well as identified additional potential humoral immune correlates. Thus, this model of infant ART and viral rebound can be used and further optimized to define biomarkers of viral rebound following long-term ART as well as to preclinically assess novel therapies to achieve a pediatric HIV functional cure.IMPORTANCE Novel interventions that do not rely on daily adherence to ART are needed to achieve sustained viral remission for perinatally infected children, who currently rely on lifelong ART. Considering the risks and expense associated with ART interruption trials, the identification of biomarkers of viral rebound will prioritize promising therapeutic intervention strategies, including anti-HIV Env protein therapeutics. However, comprehensive studies to identify those biomarkers are logistically challenging in human infants, demanding the need for relevant nonhuman primate models of HIV rebound. In this study, we developed an infant RM model of oral infection with simian-human immunodeficiency virus expressing clade C HIV Env and short-term ART followed by ATI, longitudinally characterizing the immune responses to viral infection during ART and after ATI. Additionally, we compared this infant RM model to an analogous adult RM rebound model and identified virologic and immunologic correlates of the time to viral rebound after ATI.


Subject(s)
Anti-Retroviral Agents/immunology , Anti-Retroviral Agents/pharmacology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Animals , Biomarkers , CD4-Positive T-Lymphocytes , Disease Models, Animal , Humans , Immunoglobulin G/blood , Kinetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Load , Virus Replication/drug effects , env Gene Products, Human Immunodeficiency Virus/immunology
12.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31092583

ABSTRACT

Global elimination of pediatric human immunodeficiency virus (HIV) infections will require the development of novel immune-based approaches, and understanding infant immunity to HIV is critical to guide the rational design of these intervention strategies. Despite their immunological immaturity, chronically HIV-infected children develop broadly neutralizing antibodies (bnAbs) more frequently and earlier than adults do. However, the ontogeny of humoral responses during acute HIV infection is poorly defined in infants and challenging to study in human cohorts due to the presence of maternal antibodies. To further our understanding of age-related differences in the development of HIV-specific immunity during acute infection, we evaluated the generation of virus-specific humoral immune responses in infant (n = 6) and adult (n = 12) rhesus macaques (RMs) infected with a transmitted/founder (T/F) simian-human immunodeficiency virus (SHIV) (SHIV.C.CH505 [CH505]). The plasma HIV envelope-specific IgG antibody kinetics were similar in SHIV-infected infant and adult RMs, with no significant differences in the magnitude or breadth of these responses. Interestingly, autologous tier 2 virus neutralization responses also developed with similar frequencies and kinetics in infant and adult RMs, despite infants exhibiting significantly higher follicular T helper cell (Tfh) and germinal center B cell frequencies than adults. Finally, we show that plasma viral load was the strongest predictor of the development of autologous virus neutralization in both age groups. Our results indicate that the humoral immune response to SHIV infection develops with similar kinetics among infant and adult RMs, suggesting that the early-life immune system is equipped to respond to HIV-1 and promote the production of neutralizing HIV antibodies.IMPORTANCE There is a lack of understanding of how the maturation of the infant immune system influences immunity to HIV infection or how these responses differ from those of adults. Improving our knowledge of infant HIV immunity will help guide antiviral intervention strategies that take advantage of the unique infant immune environment to successfully elicit protective immune responses. We utilized a rhesus macaque model of SHIV infection as a tool to distinguish the differences in HIV humoral immunity in infants versus adults. Here, we demonstrate that the kinetics and quality of the infant humoral immune response to HIV are highly comparable to those of adults during the early phase of infection, despite distinct differences in their Tfh responses, indicating that slightly different mechanisms may drive infant and adult humoral immunity.


Subject(s)
Age Factors , Antibody Formation , HIV Antibodies/blood , HIV Infections/immunology , HIV/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibodies, Neutralizing/blood , B-Lymphocytes/immunology , Disease Models, Animal , Germinal Center/immunology , Immunoglobulin G/blood , Macaca mulatta , Plasma/virology , T-Lymphocytes/immunology , Viral Load
13.
J Virol ; 93(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30541851

ABSTRACT

Prevention of mother-to-child transmission (MTCT) is an indispensable component in combatting the global AIDS epidemic. A combination of passive broadly neutralizing antibody (bnAb) infusion and active vaccination promises to provide protection of infants against MTCT from birth through the breastfeeding period and could prime the immune system for lifelong immunity. In this study, we investigate the impact of a single infusion of CD4 binding site (CD4bs) bnAb administered at birth on de novo antibody responses elicited by concurrent active HIV envelope vaccination. Four groups of infant macaques received active immunizations with subunit Env protein or modified vaccinia Ankara (MVA)-vectored Env and subunit Env protein, with or without a single intravenous coadministration of CH31 bnAb at birth. Vaccinated animals were monitored to evaluate binding and functional antibody responses elicited by the active vaccinations. Despite achieving plasma concentrations that were able to neutralize tier 2 viruses, coadministration of CH31 did not have a large impact on the kinetics, magnitude, specificity, or avidity of vaccine-elicited binding or functional antibody responses, including epitope specificity, the development of CD4bs antibodies, neutralization, binding to infected cells, or antibody-dependent cell-mediated cytotoxicity (ADCC). We conclude that infusion of CD4bs bnAb CH31 at birth does not interfere with de novo antibody responses to active vaccination and that a combination of passive bnAb infusion and active HIV-1 Env vaccination is a viable strategy for immediate and prolonged protection against MTCT.IMPORTANCE Our study is the first to evaluate the impact of passive infusion of a broadly neutralizing antibody in newborns on the de novo development of antibody responses following active vaccinations in infancy. We demonstrated the safety and the feasibility of bnAb administration to achieve biologically relevant levels of the antibody and showed that the passive infusion did not impair the de novo antibody production following HIV-1 Env vaccination. Our study paves the way for further investigations of the combination strategy using passive plus active immunization to provide protection of infants born to HIV-1-positive mothers over the entire period of risk for mother-to-child transmission.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/administration & dosage , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , AIDS Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Epitopes/immunology , HIV Antibodies/immunology , HIV-1/immunology , Macaca mulatta/immunology , Vaccination/methods , Vaccinia virus/genetics , Vaccinia virus/immunology
14.
AIDS Res Hum Retroviruses ; 35(3): 310-325, 2019 03.
Article in English | MEDLINE | ID: mdl-30303405

ABSTRACT

The majority of human immunodeficiency virus (HIV) type 1 infections in infants are acquired orally through breastfeeding. Toward development of a pediatric HIV vaccine to prevent breastmilk transmission, we tested the efficacy of a simultaneous oral and intramuscular (IM) vaccination regimen for preventing oral simian immunodeficiency virus (SIV) transmission in infant rhesus macaques. Two groups of neonatal macaques were immunized with DNA encoding SIV virus-like particles (DNA-SIV) on weeks 0 and 3, then boosted with modified vaccinia Ankara (MVA) virus expressing SIV antigens (MVA-SIV) on weeks 6 and 9. One group was prime/boosted by the IM route only. Another group was immunized with DNA by both the IM and topical oral (O) buccal routes, and boosted with MVA-SIV by both the IM and sublingual (SL) routes. A third group of control animals received saline by O + IM routes on weeks 0 and 3, and empty MVA by SL + IM routes on weeks 6 and 9. On week 12, infants were orally challenged once weekly with SIVmac251 until infected. The vaccine regimen that included oral routes resulted in reduced peak viremia. The rate of infection acquisition in vaccinated infants was found to be associated with prechallenge intestinal immunoglobulin G (IgG) responses to SIV gp120 and V1V2. Peak viremia was inversely correlated with postinfection intestinal IgG responses to gp120, gp41, and V1V2. These results suggest that codelivery of a pediatric HIV vaccine by an oral route may be superior to IM-only regimens for generating mucosal antibodies and preventing HIV breastmilk transmission in neonates.


Subject(s)
Membrane Glycoproteins/immunology , Mouth/virology , SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Vaccination/methods , Vaccinia virus/genetics , Viral Envelope Proteins/immunology , Administration, Oral , Animals , Animals, Newborn , Antibodies, Viral/immunology , Disease Models, Animal , HIV Infections/therapy , Immunoglobulin G/metabolism , Infectious Disease Transmission, Vertical/prevention & control , Injections, Intramuscular , Macaca mulatta , Mouth/drug effects , Mucous Membrane/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Viremia/drug therapy
15.
J Med Primatol ; 47(5): 288-297, 2018 10.
Article in English | MEDLINE | ID: mdl-30204253

ABSTRACT

BACKGROUND: A pediatric vaccine to prevent breast milk transmission of human immunodeficiency virus (HIV) may generate greater immune responses at viral entry sites if given by an oral route. METHODS: We compared immune responses induced in juvenile macaques by prime/boosting with simian immunodeficiency virus (SIV)-expressing DNA/modified vaccinia Ankara virus (MVA) by the intramuscular route (IM), the oral (O)/tonsillar routes (T), the O/sublingual (SL) routes, and O+IM/SL routes. RESULTS: O/T or O/SL immunization generated SIV-specific T cells in mucosal tissues but failed to induce SIV-specific IgA in saliva or stool or IgG in plasma. IM/IM or O+IM/SL generated humoral and cellular responses to SIV. IM/IM generated greater frequencies of TFH in spleen, but O+IM/SL animals had higher avidity plasma IgG and more often demonstrated mucosal IgA responses. CONCLUSION: These results suggest that codelivery of HIV DNA/MVA vaccines by the oral and IM routes might be optimal for generating both systemic and mucosal antibodies.


Subject(s)
Immunity, Cellular/immunology , Immunity, Mucosal/immunology , Immunogenicity, Vaccine/immunology , Macaca mulatta , Monkey Diseases/immunology , SAIDS Vaccines/adverse effects , Simian Immunodeficiency Virus/immunology , Administration, Oral , Administration, Sublingual , Animals , DNA, Viral/adverse effects , Injections, Intramuscular/adverse effects , Proof of Concept Study , Vaccinia/immunology
16.
Arthroscopy ; 34(2): 386-388, 2018 02.
Article in English | MEDLINE | ID: mdl-29413185

ABSTRACT

The ideal construct for a rotator cuff repair continues to be a hot topic for debate and biomechanical investigation. A recent study shows that anchors could be placed as close as 1 mm apart without impact on pullout strength. However, further overlap (the buddy anchor construct) led to earlier failure. This conclusion differs from other studies that have recommended the "buddy anchor" construct as a bail-out option in osteoporotic bone. Biomechanical studies are an important source of information for surgical decision making, but often do not directly predict the in vivo environment. The final determination of anchor number and placement in a rotator cuff repair should be based on 4 factors: minimizing cost, avoiding complications, using appropriate technique, and proper restoration of anatomy.


Subject(s)
Rotator Cuff/surgery , Suture Anchors , Arthroplasty , Biomechanical Phenomena , Rotator Cuff Injuries , Suture Techniques
17.
Front Immunol ; 9: 3119, 2018.
Article in English | MEDLINE | ID: mdl-30687323

ABSTRACT

Previous studies showed that single-chain fusion proteins comprised of GM-CSF and major encephalitogenic peptides of myelin, when injected subcutaneously in saline, were potent tolerogenic vaccines that suppressed experimental autoimmune encephalomyelitis (EAE) in rats and mice. These tolerogenic vaccines exhibited dominant suppressive activity in inflammatory environments even when emulsified in Complete Freund's Adjuvant (CFA). The current study provides evidence that the mechanism of tolerance was dependent upon vaccine-induced regulatory CD25+ T cells (Tregs), because treatment of mice with the Treg-depleting anti-CD25 mAb PC61 reversed tolerance. To assess tolerogenic mechanisms, we focused on 2D2-FIG mice, which have a transgenic T cell repertoire that recognizes myelin oligodendrocyte glycoprotein peptide MOG35-55 as a low-affinity ligand and the neurofilament medium peptide NFM13-37 as a high-affinity ligand. Notably, a single subcutaneous vaccination of GMCSF-MOG in saline elicited a major population of FOXP3+ Tregs that appeared within 3 days, was sustained over several weeks, expressed canonical Treg markers, and was present systemically at high frequencies in the blood, spleen, and lymph nodes. Subcutaneous and intravenous injections of GMCSF-MOG were equally effective for induction of FOXP3+ Tregs. Repeated booster vaccinations with GMCSF-MOG elicited FOXP3 expression in over 40% of all circulating T cells. Covalent linkage of GM-CSF with MOG35-55 was required for Treg induction whereas vaccination with GM-CSF and MOG35-55 as separate molecules lacked Treg-inductive activity. GMCSF-MOG elicited high levels of Tregs even when administered in immunogenic adjuvants such as CFA or Alum. Conversely, incorporation of GM-CSF and MOG35-55 as separate molecules in CFA did not support Treg induction. The ability of the vaccine to induce Tregs was dependent upon the efficiency of T cell antigen recognition, because vaccination of 2D2-FIG or OTII-FIG mice with the high-affinity ligands GMCSF-NFM or GMCSF-OVA (Ovalbumin323-339), respectively, did not elicit Tregs. Comparison of 2D2-FIG and 2D2-FIG-Rag1-/- strains revealed that GMCSF-MOG may predominantly drive Treg expansion because the kinetics of vaccine-induced Treg emergence was a function of pre-existing Treg levels. In conclusion, these findings indicate that the antigenic domain of the GMCSF-NAg tolerogenic vaccine is critical in setting the balance between regulatory and conventional T cell responses in both quiescent and inflammatory environments.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immune Tolerance/immunology , Multiple Sclerosis/therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes, Regulatory/immunology , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD4 Antigens/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Forkhead Transcription Factors/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Hybridomas , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocytosis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/genetics , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/metabolism , Vaccines/administration & dosage
18.
Clin Vaccine Immunol ; 24(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28814388

ABSTRACT

Despite success in reducing vertical HIV transmission by maternal antiretroviral therapy, several obstacles limit its efficacy during breastfeeding, and breast-milk transmission is now the dominant mode of mother-to-child transmission (MTCT) of HIV in infants. Thus, a pediatric vaccine is needed to eradicate oral HIV infections in newborns and infants. Utilizing the infant rhesus macaque model, we compared 3 different vaccine regimens: (i) HIV envelope (Env) protein only, (ii) poxvirus vector (modified vaccinia virus Ankara [MVA])-HIV Env prime and HIV Env boost, and (iii) coadministration of HIV Env and MVA-HIV Env at all time points. The vaccines were administered with an accelerated, 3-week-interval regimen starting at birth for early induction of highly functional HIV Env-specific antibodies. We also tested whether an extended, 6-week immunization interval using the same vaccine regimen as in the coadministration group would enhance the quality of antibody responses. We found that pediatric HIV vaccines administered at birth are effective in inducing HIV Env-specific plasma IgG. The vaccine regimen consisting of only HIV Env protein induced the highest levels of variable region 1 and 2 (V1V2)-specific antibodies and tier 1 neutralizing antibodies, whereas the extended-interval regimen induced both persistent Env-specific systemic IgG and mucosal IgA responses. Antibody-dependent cell-mediated cytotoxicity (ADCC) antibodies in plasma were elicited by all vaccine regimens. These data suggest that infant immunizations beginning at birth are effective for the induction of functional HIV Env-specific antibodies that could potentially protect against breast milk transmission of HIV and set the stage for immunity prior to sexual debut.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , AIDS Vaccines/administration & dosage , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibody-Dependent Cell Cytotoxicity , Genetic Vectors , HIV Envelope Protein gp120/genetics , HIV Infections/prevention & control , HIV Infections/transmission , HIV-1/immunology , Immunization, Secondary , Immunoglobulin A/blood , Infectious Disease Transmission, Vertical/prevention & control , Macaca mulatta , Models, Animal , Vaccination , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/genetics
19.
J Leukoc Biol ; 100(4): 747-760, 2016 10.
Article in English | MEDLINE | ID: mdl-27256565

ABSTRACT

Previous studies established that GM-CSF-deficient (Csf2-deficient) mice exhibit profound resistance to experimental autoimmune encephalomyelitis. This study addressed whether the resistance of Csf2-deficient mice was a result of a requirement for GM-CSF in controlling the functional balance between effector and regulatory T cell subsets during experimental autoimmune encephalomyelitis. The main observation was that treatment with the anti-CD25 mAb PC61 rendered Csf2-deficient mice fully susceptible to severe, chronic experimental autoimmune encephalomyelitis, with disease incidences and severities equivalent to that of C57BL/6 mice. When both donors and recipients were treated with PC61 in a passive model of experimental autoimmune encephalomyelitis, adoptive transfer of myelin-specific Csf2-deficient T cells into Csf2-deficient recipients resulted in a nonresolving chronic course of severe paralytic experimental autoimmune encephalomyelitis. The peripheral Csf2-deficient T cell repertoire was marked by elevated CD3+ T cell frequencies that reflected substantial accumulations of naïve CD44null-low CD4+ and CD8+ T cells but essentially normal frequencies of CD4+ CD25+ forkhead box P3+ T cells among the CD3+ T cell pool. These findings suggested that Csf2-deficient mice had secondary deficiencies in peripheral T cell sensitization to environmental antigens. In accordance, myelin oligodendrocyte glycoprotein 35-55/CFA-sensitized Csf2-deficient mice exhibited deficient peripheral sensitization to myelin oligodendrocyte glycoprotein, whereas pretreatment of Csf2-deficient mice with PC61 enabled the robust induction of myelin oligodendrocyte glycoprotein-specific T cell responses in the draining lymphatics. In conclusion, the experimental autoimmune encephalomyelitis resistance of Csf2-deficient mice, at least in part, reflects a deficient induction of effector T cell function that cannot surmount normal regulatory T cell barriers. Experimental autoimmune encephalomyelitis effector responses, however, are unleashed upon depletion of regulatory CD25+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Lymphocyte Depletion , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Disease Susceptibility , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Granulocytes/immunology , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Count , Lymphocyte Count , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology
20.
Proc Natl Acad Sci U S A ; 113(7): 1865-70, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831084

ABSTRACT

Tristetraprolin (TTP) is an inducible, tandem zinc-finger mRNA binding protein that binds to adenylate-uridylate-rich elements (AREs) in the 3'-untranslated regions (3'UTRs) of specific mRNAs, such as that encoding TNF, and increases their rates of deadenylation and turnover. Stabilization of Tnf mRNA and other cytokine transcripts in TTP-deficient mice results in the development of a profound, chronic inflammatory syndrome characterized by polyarticular arthritis, dermatitis, myeloid hyperplasia, and autoimmunity. To address the hypothesis that increasing endogenous levels of TTP in an intact animal might be beneficial in the treatment of inflammatory diseases, we generated a mouse model (TTPΔARE) in which a 136-base instability motif in the 3'UTR of TTP mRNA was deleted in the endogenous genetic locus. These mice appeared normal, but cultured fibroblasts and macrophages derived from them exhibited increased stability of the otherwise highly labile TTP mRNA. This resulted in increased TTP protein expression in LPS-stimulated macrophages and increased levels of TTP protein in mouse tissues. TTPΔARE mice were protected from collagen antibody-induced arthritis, exhibited significantly reduced inflammation in imiquimod-induced dermatitis, and were resistant to induction of experimental autoimmune encephalomyelitis, presumably by dampening the excessive production of proinflammatory mediators in all cases. These data suggest that increased systemic levels of TTP, secondary to increased stability of its mRNA throughout the body, can be protective against inflammatory disease in certain models and might be viewed as an attractive therapeutic target for the treatment of human inflammatory diseases.


Subject(s)
Inflammation/genetics , RNA, Messenger/genetics , Tristetraprolin/genetics , Aminoquinolines/adverse effects , Animals , Arthritis, Experimental/genetics , Cells, Cultured , Collagen/immunology , Dermatitis/etiology , Dermatitis/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Imiquimod , Mice , Mice, Transgenic , Mutation , Tristetraprolin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...