Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38995674

ABSTRACT

Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.


Subject(s)
Alphavirus Infections , Alphavirus , Genotype , Macaca fascicularis , RNA, Viral , Viremia , Animals , Macaca fascicularis/virology , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus Infections/virology , Alphavirus Infections/veterinary , Viremia/virology , RNA, Viral/genetics , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Phylogeny , Cytokines/genetics , Cytokines/blood
2.
Biomaterials ; 308: 122538, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38564889

ABSTRACT

Surface-enhanced Raman spectroscopy (SERS) nanotags have garnered much attention as promising bioimaging contrast agent with ultrahigh sensitivity, but their clinical translation faces challenges including biological and laser safety. As breast sentinel lymph node (SLN) imaging agents, SERS nanotags used by local injection and only accumulation in SLNs, which were removed during surgery, greatly reduce biological safety concerns. But their clinical translation lacks pilot demonstration on large animals close to humans. The laser safety requires irradiance below the maximum permissible exposure threshold, which is currently not achievable in most SERS applications. Here we report the invention of the core-shell SERS nanotags with ultrahigh brightness (1 pM limit of detection) at the second near-infrared (NIR-II) window for SLN identification on pre-clinical animal models including rabbits and non-human primate. We for the first time realize the intraoperative SERS-guided SLN navigation under a clinically safe laser (1.73 J/cm2) and identify multiple axillary SLNs on a non-human primate. No evidence of biosafety issues was observed in systematic examinations of these nanotags. Our study unveils the potential of NIR-II SERS nanotags as appropriate SLN tracers, making significant advances toward the accurate positioning of lesions using the SERS-based tracer technique.


Subject(s)
Sentinel Lymph Node , Spectrum Analysis, Raman , Animals , Spectrum Analysis, Raman/methods , Sentinel Lymph Node/diagnostic imaging , Sentinel Lymph Node/pathology , Rabbits , Female , Humans , Spectroscopy, Near-Infrared/methods
3.
Viruses ; 16(3)2024 02 27.
Article in English | MEDLINE | ID: mdl-38543734

ABSTRACT

The human immunodeficiency virus (HIV) continues to pose a significant global health challenge, with millions of people affected and new cases emerging each year. While various treatment and prevention methods exist, including antiretroviral therapy and non-vaccine approaches, developing an effective vaccine remains the most crucial and cost-effective solution to combating the HIV epidemic. Despite significant advancements in HIV research, the HIV vaccine field has faced numerous challenges, and only one clinical trial has demonstrated a modest level of efficacy. This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development using the non-human primate model (NHP) of HIV infection. NHP models offer valuable insights into potential preventive strategies for combating HIV, and they play a vital role in informing and guiding the development of novel vaccine candidates before they can proceed to human clinical trials.


Subject(s)
AIDS Vaccines , HIV Infections , HIV-1 , Animals , Humans , Primates , Vaccine Development
4.
Prog Retin Eye Res ; 100: 101247, 2024 May.
Article in English | MEDLINE | ID: mdl-38365085

ABSTRACT

Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.


Subject(s)
Disease Models, Animal , Macular Degeneration , Animals , Humans , Macular Degeneration/genetics , Macular Degeneration/physiopathology , Mice , Aging/physiology , Glaucoma/physiopathology , Glaucoma/genetics , Disease Progression
5.
Mol Ther ; 32(5): 1238-1251, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38414244

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies have demonstrated immense clinical success for B cell and plasma cell malignancies. We tested their impact on the viral reservoir in a macaque model of HIV persistence, comparing the functions of CD20 CAR T cells between animals infected with simian/human immunodeficiency virus (SHIV) and uninfected controls. We focused on the potential of this approach to disrupt B cell follicles (BCFs), exposing infected cells for immune clearance. In SHIV-infected animals, CAR T cells were highly functional, with rapid expansion and trafficking to tissue-associated viral sanctuaries, including BCFs and gut-associated lymphoid tissue (GALT). CD20 CAR T cells potently ablated BCFs and depleted lymph-node-associated follicular helper T (TFH) cells, with complete restoration of BCF architecture and TFH cells following CAR T cell contraction. BCF ablation decreased the splenic SHIV reservoir but was insufficient for effective reductions in systemic viral reservoirs. Although associated with moderate hematologic toxicity, CD20 CAR T cells were well tolerated in SHIV-infected and control animals, supporting the feasibility of this therapy in people living with HIV with underlying B cell malignancies. Our findings highlight the unique ability of CD20 CAR T cells to safely and reversibly unmask TFH cells within BCF sanctuaries, informing future combinatorial HIV cure strategies designed to augment antiviral efficacy.


Subject(s)
Antigens, CD20 , B-Lymphocytes , Disease Models, Animal , HIV Infections , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Antigens, CD20/metabolism , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Simian Immunodeficiency Virus/immunology , Immunotherapy, Adoptive/methods , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , HIV Infections/therapy , HIV Infections/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , HIV-1/immunology , Viral Load , Macaca mulatta
6.
Virol Sin ; 37(4): 610-618, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35777657

ABSTRACT

Coxsackievirus A10 (CV-A10) is one of the etiological agents associated with hand, foot and mouth disease (HFMD) and also causes a variety of illnesses in humans, including pneumonia, and myocarditis. Different people, particularly young children, may have different immunological responses to infection. Current CV-A10 infection animal models provide only a rudimentary understanding of the pathogenesis and effects of this virus. The characteristics of CV-A10 infection, replication, and shedding in humans remain unknown. In this study, rhesus macaques were infected by CV-A10 via respiratory or digestive route to mimic the HFMD in humans. The clinical symptoms, viral shedding, inflammatory response and pathologic changes were investigated in acute infection (1-11 day post infection) and recovery period (12-180 day post infection). All infected rhesus macaques during acute infection showed obvious viremia and clinical symptoms which were comparable to those observed in humans. Substantial inflammatory pathological damages were observed in multi-organs, including the lung, heart, liver, and kidney. During the acute period, all rhesus macaques displayed clinical signs, viral shedding, normalization of serum cytokines, and increased serum neutralizing antibodies, whereas inflammatory factors caused some animals to develop severe hyperglycemia during the recovery period. In addition, there were no significant differences between respiratory and digestive tract infected animals. Overall, all data presented suggest that the rhesus macaques provide the first non-human primate animal model for investigating CV-A10 pathophysiology and assessing the development of potential human therapies.


Subject(s)
Enterovirus A, Human , Hand, Foot and Mouth Disease , Animals , Antibodies, Neutralizing , Benzeneacetamides , Child, Preschool , Humans , Macaca mulatta , Piperidones
7.
Front Cell Neurosci ; 16: 889663, 2022.
Article in English | MEDLINE | ID: mdl-35602554

ABSTRACT

Retinal prostheses have shown some clinical success in patients with retinitis pigmentosa and age-related macular degeneration. However, even after the implantation of a retinal prosthesis, the patient's visual acuity is at best less than 20/420. Reduced visual acuity may be explained by a decrease in the signal-to-noise ratio due to the spontaneous hyperactivity of retinal ganglion cells (RGCs) found in degenerate retinas. Unfortunately, abnormal retinal rewiring, commonly observed in degenerate retinas, has rarely been considered for the development of retinal prostheses. The purpose of this study was to investigate the aberrant retinal network response to electrical stimulation in terms of the spatial distribution of the electrically evoked RGC population. An 8 × 8 multielectrode array was used to measure the spiking activity of the RGC population. RGC spikes were recorded in wild-type [C57BL/6J; P56 (postnatal day 56)], rd1 (P56), rd10 (P14 and P56) mice, and macaque [wild-type and drug-induced retinal degeneration (RD) model] retinas. First, we performed a spike correlation analysis between RGCs to determine RGC connectivity. No correlation was observed between RGCs in the control group, including wild-type mice, rd10 P14 mice, and wild-type macaque retinas. In contrast, for the RD group, including rd1, rd10 P56, and RD macaque retinas, RGCs, up to approximately 400-600 µm apart, were significantly correlated. Moreover, to investigate the RGC population response to electrical stimulation, the number of electrically evoked RGC spikes was measured as a function of the distance between the stimulation and recording electrodes. With an increase in the interelectrode distance, the number of electrically evoked RGC spikes decreased exponentially in the control group. In contrast, electrically evoked RGC spikes were observed throughout the retina in the RD group, regardless of the inter-electrode distance. Taken together, in the degenerate retina, a more strongly coupled retinal network resulted in the widespread distribution of electrically evoked RGC spikes. This finding could explain the low-resolution vision in prosthesis-implanted patients.

8.
J Neurodev Disord ; 14(1): 21, 2022 03 19.
Article in English | MEDLINE | ID: mdl-35305552

ABSTRACT

BACKGROUND: Fetal alcohol spectrum disorders (FASD) are common, yet preventable developmental disorders that stem from prenatal exposure to alcohol. This exposure leads to a wide array of behavioural and physical problems with a complex and poorly defined biological basis. Molecular investigations to date predominantly use rodent animal models, but because of genetic, developmental and social behavioral similarity, primate models are more relevant. We previously reported reduced cortical and hippocampal neuron levels in an Old World monkey (Chlorocebus sabaeus) model with ethanol exposure targeted to the period of rapid synaptogenesis and report here an initial molecular study of this model. The goal of this study was to evaluate mRNA expression of the hippocampus at two different behavioural stages (5 months, 2 years) corresponding to human infancy and early childhood. METHODS: Offspring of alcohol-preferring or control dams drank a maximum of 3.5 g ethanol per kg body weight or calorically matched sucrose solution 4 days per week during the last 2 months of gestation. Total mRNA expression was measured with the Affymetrix GeneChip Rhesus Macaque Genome Array in a 2 × 2 study design that interrogated two independent variables, age at sacrifice, and alcohol consumption during gestation. RESULTS AND DISCUSSION: Statistical analysis identified a preferential downregulation of expression when interrogating the factor 'alcohol' with a balanced effect of upregulation vs. downregulation for the independent variable 'age'. Functional exploration of both independent variables shows that the alcohol consumption factor generates broad functional annotation clusters that likely implicate a role for epigenetics in the observed differential expression, while the variable age reliably produced functional annotation clusters predominantly related to development. Furthermore, our data reveals a novel connection between EFNB1 and the FASDs; this is highly plausible both due to the role of EFNB1 in neuronal development as well as its central role in craniofrontal nasal syndrome (CFNS). Fold changes for key genes were subsequently confirmed via qRT-PCR. CONCLUSION: Prenatal alcohol exposure leads to global downregulation in mRNA expression. The cellular interference model of EFNB1 provides a potential clue regarding how genetically susceptible individuals may develop the phenotypic triad generally associated with classic fetal alcohol syndrome.


Subject(s)
Fetal Alcohol Spectrum Disorders , Prenatal Exposure Delayed Effects , Animals , Child, Preschool , Chlorocebus aethiops , Disease Models, Animal , Ephrin-B1/metabolism , Ephrin-B1/pharmacology , Ethanol/metabolism , Female , Fetal Alcohol Spectrum Disorders/genetics , Fetal Alcohol Spectrum Disorders/metabolism , Hippocampus/metabolism , Humans , Macaca mulatta/genetics , Macaca mulatta/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology
9.
Int J Mol Sci ; 23(5)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35269778

ABSTRACT

As in women with polycystic ovary syndrome (PCOS), hyperinsulinemia is associated with anovulation in PCOS-like female rhesus monkeys. Insulin sensitizers ameliorate hyperinsulinemia and stimulate ovulatory menstrual cycles in PCOS-like monkeys. To determine whether hyperinsulinemia (>694 pmol/L), alone, induces PCOS-like traits, five PCOS-like female rhesus monkeys with minimal PCOS-like traits, and four control females of similar mid-to-late reproductive years and body mass index, received daily subcutaneous injections of recombinant human insulin or diluent for 6−7 months. A cross-over experimental design enabled use of the same monkeys in each treatment phase. Insulin treatment unexpectedly normalized follicular phase duration in PCOS-like, but not control, females. In response to an intramuscular injection of 200 IU hCG, neither prenatally androgenized nor control females demonstrated ovarian hyperandrogenic responses while receiving insulin. An intravenous GnRH (100 ng/kg) injection also did not reveal evidence of hypergonadotropism. Taken together, these results suggest that experimentally induced adult hyperinsulinemia, alone, is insufficient to induce PCOS-like traits in female rhesus monkeys and to amplify intrinsic PCOS-like pathophysiology.


Subject(s)
Hyperandrogenism , Hyperinsulinism , Polycystic Ovary Syndrome , Animals , Female , Humans , Hyperinsulinism/chemically induced , Insulin , Macaca mulatta , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy
10.
Dev Comp Immunol ; 130: 104371, 2022 05.
Article in English | MEDLINE | ID: mdl-35131310

ABSTRACT

The short pentraxins C-reactive protein (CRP) and serum amyloid P component (SAP) are a family of pattern-recognition molecules that play versatile roles in innate immunity and inflammation. A comprehensive description is currently lacking as to the genetic characteristics of these molecules in primates. In the present study, we analyzed genetic changes of CRP and SAP genes in this phylogenic lineage. The results revealed that adaptive selection has brought about interspecific diversities of both genes. The adaptively selected amino acid changes have occurred in or adjacent to the structural domains involved in ligand- and effector-binding and homologous aggregation. Each gene, however, exhibits a striking lack of genetic variation in both commonly-used non-human primate models Macaca fascicularis and M. mulatta. These findings highlight basic facts on the genetic characteristics of primate short pentraxins and would contribute powerfully to the extrapolation of their functional insights and physiological outcomes from primate models to humans.


Subject(s)
C-Reactive Protein , Serum Amyloid P-Component , Animals , C-Reactive Protein/genetics , Inflammation , Primates , Receptors, Immunologic , Serum Amyloid P-Component/genetics
11.
Gastro Hep Adv ; 1(3): 393-402, 2022.
Article in English | MEDLINE | ID: mdl-35174366

ABSTRACT

BACKGROUND AND AIMS: Apolipoprotein A1 (A1) and haptoglobin (HP) serum levels are associated with the spread and severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We have constructed and validated a multivariable risk calculator (A1HPV6) integrating A1, HP, alpha2-macroglobulin, and gamma glutamyl transferase to improve the performances of virological biomarkers. METHODS: In a prospective observational study of hospitalized patients with nonsevere SARS-CoV-2 infection, A1HPV6 was constructed in 127 patients and validated in 116. The specificity was assessed in 7482 controls representing the general population. The primary diagnostic endpoint was the area under the receiver operating characteristic curve in patients with positive SARS-CoV-2 PCR. The primary prognostic endpoint was the age-and sex-adjusted risk of A1HPV6 to predict patients with WHO-stage > 4 (W > 4) severity. We assessed the kinetics of the A1HPV6 components in a nonhuman primate model (NHP), from baseline to 7 days (D7) after SARS-CoV-2 infection. RESULTS: The area under the receiver operating characteristic curve for A1HPV6 was 0.99 (95% CI 0.97-0.99) in the validation subset, which was not significantly different from that in the construction subset, 0.99 (0.99-0.99; P = .80), like for sensitivity 92% (85-96) vs 94% (88-97; P = .29). A1HPV6 was associated with W > 4, with a significant odds ratio of 1.3 (1.1-1.5; 0.002). In NHP, A1 levels decreased (P < .01) at D2 and normalized at D4; HP levels increased at D2 and peaked at D4. In patients, A1 concentration was very low at D2 vs controls (P < .01) and increased at D14 (P < .01) but was still lower than controls; HP increased at D2 and remained elevated at D14. CONCLUSION: These results validate the diagnostic and prognostic performances of A1HPV6. Similar kinetics of apolipoprotein A1, HP, and alpha-2-macroglobulin were observed in the NHP model. ClinicalTrials.gov number, NCT01927133.

12.
FEBS J ; 289(23): 7343-7358, 2022 12.
Article in English | MEDLINE | ID: mdl-34914205

ABSTRACT

About 40 years have passed since 'theory of mind (ToM)' research started. The false-belief test is used as a litmus test for ToM ability. The implicit false-belief test has renewed views of ToM in several disciplines, including psychology, psychiatry, and neuroscience. Many important questions have been considered via the paradigm of implicit false belief. We recently addressed the phylogenetic and physiological aspects of ToM using a version of this paradigm combined with the chemogenetic technique on Old World monkeys. We sought to create animal models for autism that exhibit behavioral phenotypes similar to human symptoms. The simultaneous manipulation of neural circuits and assessments of changes in phenotypes can help identify the causal neural substrate of ToM.


Subject(s)
Neurosciences , Theory of Mind , Humans , Phylogeny
13.
Bioengineering (Basel) ; 8(7)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34356207

ABSTRACT

The utility of implanting a bioscaffold mitral valve consisting of porcine small intestinal submucosa (PSIS) in a juvenile baboon model (12 to 14 months old at the time of implant; n = 3) to assess their in vivo tissue remodeling responses was investigated. Our findings demonstrated that the PSIS mitral valve exhibited the robust presence of de novo extracellular matrix (ECM) at all explantation time points (at 3-, 11-, and 20-months). Apart from a significantly lower level of proteoglycans in the implanted valve's annulus region (p < 0.05) at 3 months compared to the 11- and 20-month explants, there were no other significant differences (p > 0.05) found between any of the other principal valve ECM components (collagen and elastin) at the leaflet, annulus, or chordae tendinea locations, across these time points. In particular, neochordae tissue had formed, which seamlessly integrated with the native papillary muscles. However, additional processing will be required to trigger accelerated, uniform and complete valve ECM formation in the recipient. Regardless of the specific processing done to the bioscaffold valve, in this proof-of-concept study, we estimate that a 3-month window following bioscaffold valve replacement is the timeline in which complete regeneration of the valve and integration with the host needs to occur.

14.
Emerg Microbes Infect ; 10(1): 1555-1573, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34304724

ABSTRACT

To curb the pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), multiple platforms have been employed toward a safe and highly effective vaccine. Here, we develop a novel cell-based vaccine candidate, namely K562-S, by utilizing human cell K562 as a cellular carrier to display Spike (S) protein of SARS-CoV-2 on the membrane. Analogous to the traditional inactivated vaccine, K562-S cells can be propagated to a large scale by culturing and completely lose their viability after exposure to X-ray irradiation or formalin. We in turn demonstrated high immunogenicity of formalin-inactivated K562-S vaccine in both mouse and non-human primates and its protective efficacy in mice. In mice, immunization with inactivated K562-S vaccines can elicit potent neutralizing antibody (nAb) responses persisting longer than 5 months. We consequently showed in a hACE2 mouse model of SARS-CoV-2 infection that a two-shot vaccination with adjuvanted K562-S rendered greater than 3 log reduction in viral lung load and concomitant ameliorated lung pathology. Of importance, the administration of the same regimen in non-human primates was able to induce a neutralizing antibody titer averaging three-fold higher relative to human convalescent serum. These results together support the promise of K562-based, S-protein-expressing vaccines as a novel vaccination approach against SARS-CoV-2. Importantly, with a powerful capacity to carry external genes for cell-based vectors, this platform could rapidly generate two- and multiple-valent vaccines by incorporating SARS-CoV-2 mutants, SARS-CoV, or MERS-CoV.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Immunogenicity, Vaccine , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , Animals, Genetically Modified , COVID-19 Vaccines/administration & dosage , Female , HEK293 Cells , Humans , K562 Cells , Macaca mulatta , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Primates , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
15.
Stem Cell Res Ther ; 12(1): 423, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315534

ABSTRACT

BACKGROUND: Human pluripotent stem cells (hPSCs) provide a promising cell source for retinal cell replacement therapy but often lack standardized cell production and live-cell shipment logistics as well as rigorous analyses of surgical procedures for cell transplantation in the delicate macula area. We have previously established a xeno- and feeder cell-free production system for hPSC differentiated retinal pigment epithelial (RPE) cells, and herein, a novel immunosuppressed non-human primate (NHP) model with a disrupted ocular immune privilege is presented for transplanting human embryonic stem cell (hESC)-derived RPE on a scaffold, and the safety and submacular graft integration are assessed. Furthermore, the feasibility of intercontinental shipment of live hESC-RPE is examined. METHODS: Cynomolgus monkeys were systemically immunosuppressed and implanted with a hESC-RPE monolayer on a permeable polyester-terephthalate (PET) scaffold. Microscope-integrated intraoperative optical coherence tomography (miOCT)-guided surgery, postoperative follow-up incorporated scanning laser ophthalmoscopy, spectral domain (SD-) OCT, and full-field electroretinography (ERG) were used as outcome measures. In addition, histology was performed after a 28-day follow-up. RESULTS: Intercontinental cell shipment, which took >30 h from the manufacturing to the transplantation site, did not alter the hESC-RPE quality. The submacular hESC-RPE xenotransplantation was performed in 11 macaques. The miOCT typically revealed foveal disruption. ERG showed amplitude and peak time preservation in cases with favorable surgical outcomes. Histology confirmed photoreceptor preservation above the grafts and in vivo phagocytosis by hESC-RPE, albeit evidence of cytoplasmic redistribution of opsin in photoreceptors and glia hypertrophy. The immunosuppression protocol efficiently suppressed retinal T cell infiltration and microglia activation. CONCLUSION: These results suggest both structural and functional submacular integrations of hESC-RPE xenografts. It is anticipated that surgical technique refinement will further improve the engraftment of macular cell therapeutics with significant translational relevance to improve future clinical trials.


Subject(s)
Human Embryonic Stem Cells , Animals , Cell Differentiation , Cell Line , Heterografts , Humans , Primates , Retinal Pigment Epithelium , Transplantation, Heterologous
16.
Exp Neurol ; 335: 113514, 2021 01.
Article in English | MEDLINE | ID: mdl-33141071

ABSTRACT

Mild cognitive impairment is present in a number of neurodegenerative disorders including Parkinson's disease (PD). Mild cognitive impairment in PD (PD-MCI) often manifests as deficits in executive functioning, attention, and spatial and working memory. Clinical studies have suggested that the development of mild cognitive impairment may be an early symptom of PD and may even precede the onset of motor impairment by several years. Dysfunction in several neurotransmitter systems, including dopamine (DA), norepinephrine (NE), may be involved in PD-MCI, making it difficult to treat pharmacologically. In addition, many agents used to treat motor impairment in PD may exacerbate cognitive impairment. Thus, there is a significant unmet need to develop therapeutics that can treat both motor and cognitive impairments in PD. We have recently developed SK609, a selective, G-protein biased signaling agonist of dopamine D3 receptors. SK609 was successfully used to treat motor impairment and reduce levodopa-induced dyskinesia in a rodent model of PD. Further characterization of SK609 suggested that it is a selective norepinephrine transporter (NET) inhibitor with the ability to increase both DA and NE levels in the prefrontal cortex. Pharmacokinetic analysis of SK609 under systemic administration demonstrated 98% oral bioavailability and high brain distribution in striatum, hippocampus and prefrontal cortex. To evaluate the effects of SK609 on cognitive deficits of potential relevance to PD-MCI, we used unilateral 6-hydroxydopamine (6-OHDA) lesioned rats and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated cynomolgus macaques, with deficits in performance in a sustained attention and an object retrieval task, respectively. SK609 dose dependently improved the performance of 6-OHDA-lesioned rats, with peak performance achieved using a 4 mg/kg dose. This improvement was predominantly due to a significant reduction in the number of misses and false alarm errors, contributing to an increase in sustained attention. In MPTP-lesioned monkeys, this same dose also improved performance in an object retrieval task, significantly reducing cognitive errors (barrier reaches) and motor errors (fine motor dexterity problems). These data demonstrate that SK609 with its unique pharmacological effects on modulating both DA and NE can ameliorate cognitive impairment in PD models and may provide a therapeutic option to treat both motor and cognitive impairment in PD patients.


Subject(s)
Butylamines/pharmacology , Dopamine Agonists/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Parkinson Disease/drug therapy , Parkinson Disease/psychology , Psychomotor Performance/drug effects , Receptors, Dopamine D3/agonists , Animals , Attention/drug effects , Brain/metabolism , Butylamines/pharmacokinetics , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/psychology , Hydroxydopamines , MPTP Poisoning/drug therapy , Macaca fascicularis , Male , Rats , Rats, Sprague-Dawley
17.
Proc Inst Mech Eng H ; 234(9): 955-965, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32605433

ABSTRACT

Traumatic spinal cord injury is a serious neurological disorder. Patients experience a plethora of symptoms that can be attributed to the nerve fiber tracts that are compromised. This includes limb weakness, sensory impairment, and truncal instability, as well as a variety of autonomic abnormalities. This article will discuss how machine learning classification can be used to characterize the initial impairment and subsequent recovery of electromyography signals in an non-human primate model of traumatic spinal cord injury. The ultimate objective is to identify potential treatments for traumatic spinal cord injury. This work focuses specifically on finding a suitable classifier that differentiates between two distinct experimental stages (pre-and post-lesion) using electromyography signals. Eight time-domain features were extracted from the collected electromyography data. To overcome the imbalanced dataset issue, synthetic minority oversampling technique was applied. Different ML classification techniques were applied including multilayer perceptron, support vector machine, K-nearest neighbors, and radial basis function network; then their performances were compared. A confusion matrix and five other statistical metrics (sensitivity, specificity, precision, accuracy, and F-measure) were used to evaluate the performance of the generated classifiers. The results showed that the best classifier for the left- and right-side data is the multilayer perceptron with a total F-measure of 79.5% and 86.0% for the left and right sides, respectively. This work will help to build a reliable classifier that can differentiate between these two phases by utilizing some extracted time-domain electromyography features.


Subject(s)
Machine Learning , Spinal Cord Injuries , Animals , Electromyography , Humans , Primates , Spinal Cord Injuries/diagnosis , Support Vector Machine
18.
Int J Radiat Biol ; 96(1): 47-56, 2020 01.
Article in English | MEDLINE | ID: mdl-30371121

ABSTRACT

Purpose: Design and characterization of a radiation biodosimetry device are complicated by the fact that the requisite data are not available in the intended use population, namely humans exposed to a single, whole-body radiation dose. Instead, one must turn to model systems. We discuss our studies utilizing healthy, unexposed humans, human bone marrow transplant patients undergoing total body irradiation (TBI), non-human primates subjected to the same irradiation regimen received by the human TBI patients and NHPs given a single, whole-body dose of ionizing radiation.Materials and Methods: We use Bayesian linear mixed models to characterize the association between NHP and human expression patterns in radiation response genes when exposed to a common exposure regimen and across exposure regimens within the same species.Results: We show that population average differences in expression of our radiation response genes from one to another model system are comparable to typical differences between two randomly sampled members of a given model system and that these differences are smaller, on average, for linear combinations of the probe data and for the model-based combinations employed for dose prediction as part of a radiation biodosimetry device.Conclusions: Our analysis suggests that dose estimates based on our gene list will be accurate when applied to humans who have received a single, whole-body exposure to ionizing radiation.


Subject(s)
Absorption, Radiation , Animals , Bayes Theorem , Bone Marrow Transplantation , Dose-Response Relationship, Radiation , Humans , Macaca mulatta , Models, Statistical , Radiation Exposure/adverse effects , Species Specificity , Transcriptome/radiation effects
19.
J Neurotrauma ; 37(2): 410-422, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31382861

ABSTRACT

Scaling methods are used to relate animal exposure data to humans by determining equivalent biomechanical impact conditions that result in similar tissue-level mechanics for different species. However, existing scaling methods for traumatic brain injury (TBI) do not account for the anatomical and morphological complexity of the brains for different species and have not been validated based on accurate anatomy and realistic material properties. In this study, the relationship between the TBI condition and brain tissue deformation was investigated using human, baboon, and macaque brain finite element (FE) models, which featured macro- and mesoscale anatomical details. The aim was to evaluate existing scaling methods in predicting similar biomechanical responses in the different species using both idealized and real-world TBI pulses. A second aim was to develop a new method to improve how animal data are scaled to humans. As previously found in humans, the animal's brain response to the rotational head motion was well characterized by single-degree-of-freedom (sDOF) mechanical systems with resonance at certain natural frequency, and this concept was leveraged to develop a new TBI scaling method based the natural frequency of the sDOF models representing each species. Previously described biomechanical scaling methods based on mass or inertia ratios were poor predictors of equivalent strain. The novel frequency-based scaling method was an improved approach to scaling the equivalent loading conditions. The findings of this study enable better interpretation of mechanical-trauma responses obtained from animal data to the human, thus effectively advancing the development of human injury criteria and contributing toward the mitigation of TBI.


Subject(s)
Brain Injuries, Traumatic/diagnosis , Brain/pathology , Animals , Biomechanical Phenomena , Brain/physiopathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Finite Element Analysis , Humans , Injury Severity Score , Macaca , Papio
20.
Int J Mol Sci ; 20(24)2019 Dec 07.
Article in English | MEDLINE | ID: mdl-31817864

ABSTRACT

Vascular cognitive impairment (VCI) or vascular dementia occurs as a result of brain ischemia and represents the second most common type of dementia after Alzheimer's disease. To explore the underlying mechanisms of VCI, several animal models of chronic cerebral hypoperfusion have been developed in rats, mice, and primates. We established a mouse model of chronic cerebral hypoperfusion by narrowing the bilateral common carotid arteries with microcoils, eventually resulting in hippocampal atrophy. In addition, a mouse model of white matter infarct-related damage with cognitive and motor dysfunction has also been established by asymmetric common carotid artery surgery. Although most experiments studying chronic cerebral hypoperfusion have been performed in rodents because of the ease of handling and greater ethical acceptability, non-human primates appear to represent the best model for the study of VCI, due to their similarities in much larger white matter volume and amyloid ß depositions like humans. Therefore, we also recently developed a baboon model of VCI through three-vessel occlusion (both the internal carotid arteries and the left vertebral artery). In this review, several animal models of chronic cerebral hypoperfusion, from mouse to primate, are extensively discussed to aid in better understanding of pathophysiology of VCI.


Subject(s)
Brain Ischemia/complications , Cognition Disorders/pathology , Dementia, Vascular/pathology , Disease Models, Animal , Animals , Chronic Disease , Cognition Disorders/etiology , Dementia, Vascular/etiology , Mice , Primates
SELECTION OF CITATIONS
SEARCH DETAIL