Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell Mol Gastroenterol Hepatol ; 18(1): 53-70, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38438014

ABSTRACT

BACKGROUND & AIMS: The abdominal discomfort experienced by patients with colitis may be attributable in part to the presence of small intestinal dysmotility, yet mechanisms linking colonic inflammation with small-bowel motility remain largely unexplored. We hypothesize that colitis results in small intestinal hypomotility owing to a loss of enteroendocrine cells (EECs) within the small intestine that can be rescued using serotonergic-modulating agents. METHODS: Male C57BL/6J mice, as well as mice that overexpress (EECOVER) or lack (EECDEL) NeuroD1+ enteroendocrine cells, were exposed to dextran sulfate sodium (DSS) colitis (2.5% or 5% for 7 days) and small intestinal motility was assessed by 70-kilodalton fluorescein isothiocyanate-dextran fluorescence transit. EEC number and differentiation were evaluated by immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining, and quantitative reverse-transcriptase polymerase chain reaction. Mice were treated with the 5-hydroxytryptamine receptor 4 agonist prucalopride (5 mg/kg orally, daily) to restore serotonin signaling. RESULTS: DSS-induced colitis was associated with a significant small-bowel hypomotility that developed in the absence of significant inflammation in the small intestine and was associated with a significant reduction in EEC density. EEC loss occurred in conjunction with alterations in the expression of key serotonin synthesis and transporter genes, including Tph1, Ddc, and Slc6a4. Importantly, mice overexpressing EECs revealed improved small intestinal motility, whereas mice lacking EECs had worse intestinal motility when exposed to DSS. Finally, treatment of DSS-exposed mice with the 5-hydroxytryptamine receptor 4 agonist prucalopride restored small intestinal motility and attenuated colitis. CONCLUSIONS: Experimental DSS colitis induces significant small-bowel dysmotility in mice owing to enteroendocrine loss that can be reversed by genetic modulation of EEC or administering serotonin analogs, suggesting novel therapeutic approaches for patients with symptomatic colitis.

2.
Gut Microbes ; 15(1): 2221470, 2023.
Article in English | MEDLINE | ID: mdl-37312412

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating disease in premature infants and the leading cause of death and disability from gastrointestinal disease in this vulnerable population. Although the pathophysiology of NEC remains incompletely understood, current thinking indicates that the disease develops in response to dietary and bacterial factors in the setting of a vulnerable host. As NEC progresses, intestinal perforation can result in serious infection with the development of overwhelming sepsis. In seeking to understand the mechanisms by which bacterial signaling on the intestinal epithelium can lead to NEC, we have shown that the gram-negative bacterial receptor toll-like receptor 4 is a critical regulator of NEC development, a finding that has been confirmed by many other groups. This review article provides recent findings on the interaction of microbial signaling, the immature immune system, intestinal ischemia, and systemic inflammation in the pathogenesis of NEC and the development of sepsis. We will also review promising therapeutic approaches that show efficacy in pre-clinical studies.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Newborn, Diseases , Sepsis , Infant , Infant, Newborn , Humans , Infant, Premature
3.
Am J Physiol Gastrointest Liver Physiol ; 325(1): G23-G41, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37120853

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.


Subject(s)
Brain Injuries , Cognitive Dysfunction , Enterocolitis, Necrotizing , Humans , Infant, Newborn , Infant , Female , Animals , Mice , Milk, Human/metabolism , Brain-Derived Neurotrophic Factor , Neuroinflammatory Diseases , Enterocolitis, Necrotizing/etiology , Oligosaccharides/pharmacology , Oligosaccharides/therapeutic use , Oligosaccharides/analysis , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/complications , Brain Injuries/complications , Brain Injuries/metabolism
4.
Semin Perinatol ; 47(1): 151695, 2023 02.
Article in English | MEDLINE | ID: mdl-36599763

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of death and disability from gastrointestinal disease in premature infants. The mortality of patients with NEC is approximately 30%, a figure that has not changed in many decades, reflecting the need for a greater understanding of its pathogenesis. Progress towards understanding the cellular and molecular mechanisms underlying NEC requires the study of highly translational animal models. Such animal models must mimic the biology and physiology of premature infants, while still allowing for safe experimental manipulation of environmental and microbial factors thought to be associated with the risk and severity of NEC. Findings from animal models have yielded insights into the interactions between the host, the colonizing microbes, and the innate immune receptor Toll-like Receptor 4 (TLR4) in driving disease development. This review discusses the relative strengths and weaknesses of available in vivo, in vitro, and NEC-in-a-dish models of this disease. We also highlight the unique contributions that each model has made to our understanding of the complex interactions between enterocytes, microbiota, and immune cells in the pathogenesis of NEC. The overall purpose of this review is to provide a menu of options regarding currently available animal models of NEC, while in parallel hopefully reducing the potential uncertainty and confusion regarding NEC models to assist those who wish to enter this field from other disciplines.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Microbiota , Animals , Female , Infant, Newborn , Humans , Infant, Premature , Models, Animal , Disease Models, Animal
5.
Am J Physiol Gastrointest Liver Physiol ; 323(3): G265-G282, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35819175

ABSTRACT

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and is steadily rising in frequency. Patients who develop NEC have a very high mortality, illustrating the importance of developing novel prevention or treatment approaches. We and others have shown that NEC arises in part from exaggerated signaling via the bacterial receptor, Toll-like receptor 4 (TLR4) on the intestinal epithelium, leading to widespread intestinal inflammation and intestinal ischemia. Strategies that limit the extent of TLR4 signaling, including the administration of amniotic fluid, can reduce NEC development in mouse and piglet models. We now seek to test the hypothesis that a secretome derived from amnion-derived cells can prevent or treat NEC in preclinical models of this disease via a process involving TLR4 inhibition. In support of this hypothesis, we show that the administration of this secretome, named ST266, to mice or piglets can prevent and treat experimental NEC. The protective effects of ST266 occurred in the presence of marked TLR4 inhibition in the intestinal epithelium of cultured epithelial cells, intestinal organoids, and human intestinal samples ex vivo, independent of epidermal growth factor. Strikingly, RNA-seq analysis of the intestinal epithelium in mice reveals that the ST266 upregulates critical genes associated with gut remodeling, intestinal immunity, gut differentiation. and energy metabolism. These findings show that the amnion-derived secretome ST266 can prevent and treat NEC, suggesting the possibility of novel therapeutic approaches for patients with this devastating disease.NEW & NOTEWORTHY This work provides hope for children who develop NEC, a devastating disease of premature infants that is often fatal, by revealing that the secreted product of amniotic progenitor cells (called ST266) can prevent or treat NEC in mice, piglet, and "NEC-in-a-dish" models of this disease. Mechanistically, ST266 prevented bacterial signaling, and a detailed transcriptomic analysis revealed effects on gut differentiation, immunity, and metabolism. Thus, an amniotic secretome may offer novel approaches for NEC.


Subject(s)
Enterocolitis, Necrotizing , Multipotent Stem Cells , Secretome , Amnion/cytology , Animals , Disease Models, Animal , Enterocolitis, Necrotizing/prevention & control , Intestinal Mucosa/metabolism , Mice , Multipotent Stem Cells/metabolism , Swine , Toll-Like Receptor 4/metabolism
6.
Sci Transl Med ; 13(612): eabg3459, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34550727

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating disease of premature infants, whose pathogenesis remains incompletely understood, although activation of the Gram-negative bacterial receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium plays a critical role. Patients with NEC typically display gastrointestinal dysmotility before systemic disease is manifest, suggesting that dysmotility could drive NEC development. Both intestinal motility and inflammation are governed by the enteric nervous system, a network of enteric neurons and glia. We hypothesized here that enteric glia loss in the premature intestine could lead to dysmotility, exaggerated TLR4 signaling, and NEC development. We found that intestinal motility is reduced early in NEC in mice, preceding the onset of intestinal inflammation, whereas pharmacologic restoration of intestinal motility reduced NEC severity. Ileal samples from mouse, piglet, and human NEC revealed enteric glia depletion, and glia-deficient mice (Plp1ΔDTR, Sox10ΔDTR, and BdnfΔDTR) showed increased NEC severity compared with wild-type mice. Mice lacking TLR4 on enteric glia (Sox10-Tlr4ko) did not show NEC-induced enteric glia depletion and were protected from NEC. Mechanistically, brain-derived neurotrophic factor (BDNF) from enteric glia restrained TLR4 signaling on the intestine to prevent NEC. BDNF was reduced in mouse and human NEC, and BDNF administration reduced both TLR4 signaling and NEC severity in enteric glia­deficient mice. Last, we identified an agent (J11) that enhanced enteric glial BDNF release, inhibited intestinal TLR4, restored motility, and prevented NEC in mice. Thus, enteric glia loss might contribute to NEC through intestinal dysmotility and increased TLR4 activation, suggesting enteric glia therapies for this disorder.


Subject(s)
Enterocolitis, Necrotizing , Toll-Like Receptor 4 , Humans , Infant, Newborn , Neuroglia , Toll-Like Receptor 4/genetics , Animals , Mice
7.
Front Immunol ; 12: 650709, 2021.
Article in English | MEDLINE | ID: mdl-33968047

ABSTRACT

Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.


Subject(s)
Enterocolitis, Necrotizing/immunology , Gastrointestinal Microbiome/immunology , Infant, Newborn, Diseases/immunology , Prenatal Exposure Delayed Effects/immunology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Infant, Very Low Birth Weight/immunology , Pregnancy
8.
Nat Commun ; 12(1): 1042, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589625

ABSTRACT

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Enterocolitis, Necrotizing/genetics , Indoles/administration & dosage , Milk, Human/physiology , Receptors, Aryl Hydrocarbon/genetics , Toll-Like Receptor 4/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/immunology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/immunology , Diet/methods , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/prevention & control , Female , Gene Expression Regulation , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Ligands , Maternal Exposure , Mice , Pregnancy , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , Swine , Toll-Like Receptor 4/immunology
9.
Eur J Surg Oncol ; 47(6): 1278-1285, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33500181

ABSTRACT

BACKGROUND: A rare appendiceal malignancy is characterized by both glandular and neuroendocrine histology. It often presents with dissemination of the perforated tumor to peritoneal surfaces. Current treatments involve systemic chemotherapy, cytoreductive surgery and perioperative intraperitoneal chemotherapy. METHODS: The impact of clinical, histological and treatment-related characteristics on survival were evaluated and subjected to univariate statistical analyses. All patients had stage IV disease and were treated by a uniform treatment strategy. Survival was determined from onset of disease until death or most recent follow-up. RESULTS: There were 47 patients available for study of whom 17 were male. Median age was 48 with a range of 27-65. None or a single symptom vs. 2 or more symptoms had a significant effect on survival. Median survival of the entire cohort was 45 months and 34.88% and 8.72% of patients survived 5 and 10 years, respectively. The use of neoadjuvant chemotherapy showed no impact on survival. Patients with a peritoneal cancer index (PCI) of 0-20 as compared to PCI > 20 survived longer (p = 0.012). The survival of patients able to have a complete resection as compared to an incomplete resection of disease was significant (p = 0.0087). The type of perioperative chemotherapy did not alter survival. CONCLUSIONS: These data show that patients with a lesser extent of disease with a complete cytoreduction had an improved prognosis. No benefit from systemic or perioperative regional chemotherapy was apparent. With long-term follow-up, patients with the combined glandular and neuroendocrine histology exhibiting peritoneal metastases have a guarded prognosis.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Appendiceal Neoplasms/therapy , Carcinoid Tumor/therapy , Neoplasms, Complex and Mixed/therapy , Peritoneal Neoplasms/therapy , Adenocarcinoma/complications , Adenocarcinoma/secondary , Administration, Intravenous , Adult , Aged , Appendiceal Neoplasms/complications , Appendiceal Neoplasms/pathology , Carcinoid Tumor/complications , Carcinoid Tumor/secondary , Cytoreduction Surgical Procedures , Doxorubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Hyperthermic Intraperitoneal Chemotherapy , Infusions, Parenteral , Leucovorin/administration & dosage , Male , Middle Aged , Mitomycin/administration & dosage , Neoadjuvant Therapy , Neoplasm Staging , Neoplasm, Residual , Neoplasms, Complex and Mixed/pathology , Neuroendocrine Tumors , Perioperative Period , Peritoneal Neoplasms/complications , Peritoneal Neoplasms/secondary , Prognosis , Survival Rate , Symptom Assessment
10.
Front Immunol ; 11: 899, 2020.
Article in English | MEDLINE | ID: mdl-32499778

ABSTRACT

Many functions of the immune system are impaired in neonates, allowing vulnerability to serious bacterial, viral and fungal infections which would otherwise not be pathogenic to mature individuals. This vulnerability is exacerbated in compromised newborns such as premature neonates and those who have undergone surgery or who require care in an intensive care unit. Higher susceptibility of preterm neonates to infections is associated with delayed immune system maturation, with deficiencies present in both the innate and adaptive immune components. Here, we review recent insights into early life immunity, and highlight features associated with compromised newborns, given the challenges of studying neonatal immunity in compromised neonates due to the transient nature of this period of life, and logistical and ethical obstacles posed by undertaking studies newborns and infants. Finally, we highlight how the unique immunological characteristics of the premature host play key roles in the pathogenesis of diseases that are unique to this population, including necrotizing enterocolitis and the associated sequalae of lung and brain injury.


Subject(s)
Immunity, Mucosal , Infant, Premature/immunology , Enterocolitis, Necrotizing/etiology , Humans , Infant, Newborn
11.
Surg Oncol ; 29: 20-24, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31196489

ABSTRACT

BACKGROUND: Minimally invasive surgeries are increasingly being performed for primary colon cancer resections since laparoscopic and robotic surgeries have less post-operative pain, shorter length of hospitalization, less morbidity, improved patient satisfaction and equivalent R0 resection rates compared to laparotomy. METHODS: To analyze characteristics of patients who developed port site metastases after minimally invasive colectomy, a retrospective case series of a single institution from 2004 to 2017 was performed. The study included patients who had a minimally invasive resection of the primary colon cancer and subsequent cytoreduction and heated intraperitoneal chemotherapy (CRS/HIPEC) for peritoneal metastases. Patient characteristics, histology, pathology, prior treatments, time between surgeries, carcinoembryonic antigen (CEA) levels and survival were reviewed. RESULTS: There were 123 patients who had CRS/HIPEC and 13 of them had a history of laparoscopic or robotic colectomy followed by the development of port site disease. Four were females, nine were males. Median age was 48 years (range, 19-64). Eleven of 13 primary colon cancers were T3 or T4. Ten of 13 patients had no clinical evidence of peritoneal metastases at the time of initial resection. All 13 patients had metastatic deposits at port sites that were confirmed histopathologically at the time of CRS/HIPEC. CONCLUSIONS: Port site metastases were present concomitantly with peritoneal metastases in 13 patients. An advanced T-stage of disease occurred in 85% of patients. Port site metastases do occur after minimally invasive colon resection.


Subject(s)
Colorectal Neoplasms/surgery , Laparoscopy/adverse effects , Minimally Invasive Surgical Procedures/adverse effects , Peritoneal Neoplasms/epidemiology , Peritoneal Neoplasms/secondary , Postoperative Complications , Adult , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Young Adult
12.
J Immigr Minor Health ; 18(6): 1301-1308, 2016 12.
Article in English | MEDLINE | ID: mdl-26429573

ABSTRACT

Cardiovascular disease (CVD) remains the leading cause of death in the United States (US). African-descent populations bear a disproportionate burden of CVD risk factors. With the increase in the number of West African immigrants (WAIs) to the US over the past decades, it is imperative to specifically study this new and substantial subset of the African-descent population and how acculturation impacts their CVD risk. The Afro-Cardiac study, a community-based cross-sectional study of adult WAIs in the Baltimore-Washington metropolis. Guided by the PRECEDE-PROCEED model, we used a modification of the World Health Organization Steps survey to collect data on demographics, socioeconomic status, migration-related factors and behaviors. We obtained physical, biochemical, acculturation measurements as well as a socio-demographic and health history. Our study provides critical data on the CVD risk of WAIs. The framework used is valuable for future epidemiological studies addressing CVD risk and acculturation among immigrants.


Subject(s)
Acculturation , Black or African American/statistics & numerical data , Cardiovascular Diseases/ethnology , Emigrants and Immigrants/statistics & numerical data , Adult , Africa, Western/ethnology , Aged , Blood Glucose , Body Mass Index , Cross-Sectional Studies , Female , Health Behavior , Humans , Hypertension/ethnology , Lipids/blood , Male , Middle Aged , Prevalence , Research Design , Risk Factors , Self Report , Socioeconomic Factors , United States/epidemiology
13.
BMJ Case Rep ; 20152015 Jul 06.
Article in English | MEDLINE | ID: mdl-26150637

ABSTRACT

A 67-year-old man presented to the emergency department with chronic weakness, fatigue and failure to thrive. On physical examination, he was found to have multifocal exophytic cutaneous masses in the pubic and scrotal regions. We obtained a shave biopsy, and subsequent histopathology demonstrated non-native tissue consistent with metastasis from a primary adenocarcinoma. We report this novel case of anogenital cutaneous metastases of colorectal adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Anus Neoplasms/secondary , Colonic Neoplasms/pathology , Skin Neoplasms/secondary , Skin/pathology , Urogenital Neoplasms/secondary , Aged , Biopsy , Humans , Intestine, Large/pathology , Male , Scrotum/pathology
14.
AIDS ; 29(10): 1137-46, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26035316

ABSTRACT

OBJECTIVE: Several observational studies suggest that medroxyprogesterone acetate (MPA) injectable contraceptives may increase a woman's risk of sexual HIV-1 acquisition. In-vitro studies are conflicting, mainly due to differences in the type of progestin studied or activation status of the primary cells. We sought to determine whether MPA increases infection of unstimulated peripheral blood mononuclear cells (PBMCs). METHODS: Freshly isolated PBMCs from normal blood donors were treated with physiologic MPA concentrations ranging from 0.003 to 5 ng/ml and infected with GFP-tagged R5-tropic or X4-tropic HIV-1 pseudoviruses by spinoculation. The infection was limited to a single cycle. Cells were stained with CD3, CD8 and CD14. Infection was quantified as the percentage of GFP cells by flow cytometry. RESULTS: Absolute infection was greater among unstimulated MPA-treated CD3⁺CD8⁻ T cells vs. untreated cells across MPA concentrations of 0.003-3 ng/ml using R5 (P < 0.003) and 0.03-0.3 ng/ml using X4 (P < 0.005) pseudovirus. There was increased relative infection of CD3⁺CD8⁻ T cells in MPA-treated whole PBMC cultures but not after monocytes were depleted (P < 0.02). HIV-1 infection of stimulated PBMC showed no differences in R5 or X4 infection across all MPA concentrations (P > 0.5). CONCLUSION: The CD3⁺CD8⁻ T-cell population of MPA-treated unstimulated PBMCs were more susceptible to HIV-1 infection than untreated cells. The increased infection was partly due to monocytes and was lost when PBMC were exogenously stimulated. These data provide confirmation of a biological association between MPA exposure and increased susceptibility to HIV-1 infection, particularly among women who inject drugs.


Subject(s)
Contraceptive Agents/metabolism , HIV-1/isolation & purification , Immunologic Factors/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Medroxyprogesterone Acetate/metabolism , CD3 Complex/analysis , CD8 Antigens/analysis , Female , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins/analysis , Humans , Lipopolysaccharide Receptors/analysis , Male
15.
Nat Med ; 18(3): 446-51, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344296

ABSTRACT

Highly active antiretroviral therapy (HAART) has dramatically decreased mortality from HIV-1 infection and is a major achievement of modern medicine. However, there is no fundamental theory of HAART. Elegant models describe the dynamics of viral replication, but a metric for the antiviral activity of drug combinations relative to a target value needed for control of replication is lacking. Treatment guidelines are based on empirical results of clinical trials in which other factors such as regimen tolerability also affect outcome. Why only certain drug combinations control viral replication remains unclear. Here we quantify the intrinsic antiviral activity of antiretroviral drug combinations. We show that most single antiretroviral drugs show previously unappreciated complex nonlinear pharmacodynamics that determine their inhibitory potential at clinical concentrations. We demonstrate that neither of the major theories for drug combinations accurately predicts the combined effects of multiple antiretrovirals. However, the combined effects can be understood with a new approach that considers the degree of independence of drug effects. This analysis allows a direct comparison of the inhibitory potential of different drug combinations under clinical concentrations, reconciles the results of clinical trials, defines a target level of inhibition associated with treatment success and provides a rational basis for treatment simplification and optimization.


Subject(s)
Anti-Retroviral Agents/pharmacokinetics , Antiretroviral Therapy, Highly Active/standards , Drug Combinations , Drug Therapy, Combination/standards , HIV Infections/drug therapy , Algorithms , Anti-Retroviral Agents/standards , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active/methods , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , HIV-1/pathogenicity , Humans , Virus Replication
16.
Proc Natl Acad Sci U S A ; 108(18): 7613-8, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502494

ABSTRACT

HIV-1 drug resistance is a major clinical problem. Resistance is evaluated using in vitro assays measuring the fold change in IC(50) caused by resistance mutations. Antiretroviral drugs are used at concentrations above IC(50), however, and inhibition at clinical concentrations can only be predicted from IC(50) if the shape of the dose-response curve is also known. Curve shape is influenced by cooperative interactions and is described mathematically by the slope parameter or Hill coefficient (m). Implicit in current analysis of resistance is the assumption that mutations shift dose-response curves to the right without affecting the slope. We show here that m is altered by resistance mutations. For reverse transcriptase and fusion inhibitors, single resistance mutations affect both slope and IC(50). For protease inhibitors, single mutations primarily affect slope. For integrase inhibitors, only IC(50) is affected. Thus, there are fundamental pharmacodynamic differences in resistance to different drug classes. Instantaneous inhibitory potential (IIP), the log inhibition of single-round infectivity at clinical concentrations, takes into account both slope and IC(50), and thus provides a direct measure of the reduction in susceptibility produced by mutations and the residual activity of drugs against resistant viruses. The standard measure, fold change in IC(50), does not correlate well with changes in IIP when mutations alter slope. These results challenge a fundamental assumption underlying current analysis of HIV-1 drug resistance and suggest that a more complete understanding of how resistance mutations reduce antiviral activity requires consideration of a previously ignored parameter, the dose-response curve slope.


Subject(s)
Dose-Response Relationship, Drug , Drug Resistance, Viral/genetics , HIV Fusion Inhibitors/metabolism , HIV Integrase Inhibitors/metabolism , HIV Protease Inhibitors/metabolism , HIV-1/genetics , Inhibitory Concentration 50 , Cell Line , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HIV Fusion Inhibitors/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Protease Inhibitors/pharmacology , Humans , Models, Biological , Mutation/genetics , Regression Analysis
17.
AIDS Res Hum Retroviruses ; 27(2): 183-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20919920

ABSTRACT

Elite controllers or suppressors (ES) are HIV-1-infected patients who maintain viral loads of <50 copies/ml without antiretroviral therapy. While HLA-B*57 and B*5801 alleles are overrepresented in ES, many HLA-B*57/B*5801 patients become chronic progressors (CP). We show here that HIV-1 infection results in similar levels of downregulation of HLA-B*57 and HLA-B*5801 molecules on primary CD4(+) T cells from ES and CP. Thus, differences in HIV-1-mediated downregulation of HLA-B*57/B*5801 molecules do not distinguish ES from CP.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Down-Regulation , HIV-1/physiology , HLA-B Antigens/metabolism , Alleles , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...