Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Neuroimmune Pharmacol ; 19(1): 28, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862787

ABSTRACT

Despite antiretroviral therapy (ART), HIV-associated peripheral neuropathy remains one of the most prevalent neurologic manifestations of HIV infection. The spinal cord is an essential component of sensory pathways, but spinal cord sampling and evaluation in people with HIV has been very limited, especially in those on ART. The SIV/macaque model allows for assessment of the spinal cord at key time points throughout infection with and without ART. In this study, RNA was isolated from the spinal cord of uninfected, SIV+, and SIV + ART animals to track alterations in gene expression using global RNA-seq. Next, the SeqSeek platform was used to map changes in gene expression to specific cell types. Pathway analysis of differentially expressed genes demonstrated that highly upregulated genes in SIV-infected spinal cord aligned with interferon and viral response pathways. Additionally, this upregulated gene set significantly overlapped with those expressed in myeloid-derived cells including microglia. Downregulated genes were involved in cholesterol and collagen biosynthesis, and TGF-b regulation of extracellular matrix. In contrast, enriched pathways identified in SIV + ART animals included neurotransmitter receptors and post synaptic signaling regulators, and transmission across chemical synapses. SeqSeek analysis showed that upregulated genes were primarily expressed by neurons rather than glia. These findings indicate that pathways activated in the spinal cord of SIV + ART macaques are predominantly involved in neuronal signaling rather than proinflammatory pathways. This study provides the basis for further evaluation of mechanisms of SIV infection + ART within the spinal cord with a focus on therapeutic interventions to maintain synaptodendritic homeostasis.


Subject(s)
Neuroglia , Neurons , Simian Acquired Immunodeficiency Syndrome , Spinal Cord , Animals , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/drug therapy , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/virology , Neuroglia/metabolism , Neuroglia/drug effects , Neuroglia/virology , Neurons/metabolism , Neurons/drug effects , Neurons/virology , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/pharmacology , Simian Immunodeficiency Virus/drug effects , Macaca mulatta , Gene Expression/drug effects , Male , Gene Expression Regulation/drug effects
2.
Nat Commun ; 15(1): 3738, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702297

ABSTRACT

Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.


Subject(s)
Aluminum Hydroxide , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccines, Inactivated , Animals , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Female , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Mice , Vaccines, Inactivated/immunology , SARS-CoV-2/immunology , Aluminum Hydroxide/administration & dosage , Disease Models, Animal , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine , Antibodies, Viral/immunology , Mice, Inbred BALB C , Humans , Severe acute respiratory syndrome-related coronavirus/immunology
3.
Nat Commun ; 15(1): 4606, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816375

ABSTRACT

Our limited understanding of the pathophysiological mechanisms that operate during sepsis is an obstacle to rational treatment and clinical trial design. There is a critical lack of data from low- and middle-income countries where the sepsis burden is increased which inhibits generalized strategies for therapeutic intervention. Here we perform RNA sequencing of whole blood to investigate longitudinal host response to sepsis in a Ghanaian cohort. Data dimensional reduction reveals dynamic gene expression patterns that describe cell type-specific molecular phenotypes including a dysregulated myeloid compartment shared between sepsis and COVID-19. The gene expression signatures reported here define a landscape of host response to sepsis that supports interventions via targeting immunophenotypes to improve outcomes.


Subject(s)
COVID-19 , Phenotype , Sepsis , Transcriptome , Humans , Sepsis/genetics , Sepsis/blood , Sepsis/immunology , COVID-19/immunology , COVID-19/genetics , COVID-19/blood , COVID-19/virology , Ghana/epidemiology , Male , Cohort Studies , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Female , Adult , Middle Aged , Gene Expression Profiling , Sequence Analysis, RNA
4.
Res Sq ; 2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37961507

ABSTRACT

Inactivated whole virus SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide (Alum) are among the most widely used COVID-19 vaccines globally and have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous virus infection in healthy recipients, the emergence of novel SARS-CoV-2 variants and the presence of large zoonotic reservoirs provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes including vaccine-associated enhanced respiratory disease (VAERD). To evaluate this possibility, we tested the performance of an inactivated SARS-CoV-2 vaccine (iCoV2) in combination with Alum against either homologous or heterologous coronavirus challenge in a mouse model of coronavirus-induced pulmonary disease. Consistent with human results, iCoV2 + Alum protected against homologous challenge. However, challenge with a heterologous SARS-related coronavirus, Rs-SHC014-CoV (SHC014), up to at least 10 months post-vaccination, resulted in VAERD in iCoV2 + Alum-vaccinated animals, characterized by pulmonary eosinophilic infiltrates, enhanced pulmonary pathology, delayed viral clearance, and decreased pulmonary function. In contrast, vaccination with iCoV2 in combination with an alternative adjuvant (RIBI) did not induce VAERD and promoted enhanced SHC014 clearance. Further characterization of iCoV2 + Alum-induced immunity suggested that CD4+ T cells were a major driver of VAERD, and these responses were partially reversed by re-boosting with recombinant Spike protein + RIBI adjuvant. These results highlight potential risks associated with vaccine breakthrough in recipients of Alum-adjuvanted inactivated vaccines and provide important insights into factors affecting both the safety and efficacy of coronavirus vaccines in the face of heterologous virus infections.

5.
Viruses ; 15(5)2023 04 26.
Article in English | MEDLINE | ID: mdl-37243143

ABSTRACT

The alphavirus chikungunya virus (CHIKV) represents a reemerging public health threat as mosquito vectors spread and viruses acquire advantageous mutations. Although primarily arthritogenic in nature, CHIKV can produce neurological disease with long-lasting sequelae that are difficult to study in humans. We therefore evaluated immunocompetent mouse strains/stocks for their susceptibility to intracranial infection with three different CHIKV strains, the East/Central/South African (ECSA) lineage strain SL15649 and Asian lineage strains AF15561 and SM2013. In CD-1 mice, neurovirulence was age- and CHIKV strain-specific, with SM2013 inducing less severe disease than SL15649 and AF15561. In 4-6-week-old C57BL/6J mice, SL15649 induced more severe disease and increased viral brain and spinal cord titers compared to Asian lineage strains, further indicating that neurological disease severity is CHIKV-strain-dependent. Proinflammatory cytokine gene expression and CD4+ T cell infiltration in the brain were also increased with SL15649 infection, suggesting that like other encephalitic alphaviruses and with CHIKV-induced arthritis, the immune response contributes to CHIKV-induced neurological disease. Finally, this study helps overcome a current barrier in the alphavirus field by identifying both 4-6-week-old CD-1 and C57BL/6J mice as immunocompetent, neurodevelopmentally appropriate mouse models that can be used to examine CHIKV neuropathogenesis and immunopathogenesis following direct brain infection.


Subject(s)
Chikungunya Fever , Chikungunya virus , Encephalomyelitis , Humans , Mice , Animals , Mice, Inbred C57BL , Chikungunya virus/physiology , Virus Replication
6.
Comp Med ; 71(5): 383-397, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34610856

ABSTRACT

Mice are an invaluable resource for studying virus-induced disease. They are a small, genetically modifiable animal for which a large arsenal of genetic and immunologic tools is available for evaluation of pathogenesis and potential vaccines and therapeutics. SARS-CoV-2, the betacoronavirus responsible for the COVID-19 pandemic, does not naturally replicate in wild-type mice, due to structural differences between human and mouse ACE2, the primary receptor for SARS-CoV-2 entry into cells. However, several mouse strains have been developed that allow for SARS-CoV-2 replication and clinical disease. Two broad strategies have primarily been deployed for developing mouse strains susceptible to COVID-19-like disease: adding in the human ACE2 gene and adapting the virus to the mouse ACE2 receptor. Both approaches result in mice that develop several of the clinical and pathologic hallmarks of COVID-19, including acute respiratory distress syndrome and acute lung injury. In this review, we describe key acute pulmonary and extrapulmonary pathologic changes seen in COVID-19 patients that mouse models of SARS-CoV-2 infection ideally replicate, the essential development of mouse models for the study of Severe Acute Respiratory Syndrome and Middle Eastern Respiratory Syndrome and the basis of many of the models of COVID-19, and key clinical and pathologic features of currently available mouse models of SARS-CoV-2 infection.


Subject(s)
COVID-19 , Pandemics , Animals , Disease Models, Animal , Humans , Lung , Mice , SARS-CoV-2
7.
PLoS Pathog ; 17(9): e1009897, 2021 09.
Article in English | MEDLINE | ID: mdl-34492082

ABSTRACT

The key to battling the COVID-19 pandemic and its potential aftermath is to develop a variety of vaccines that are efficacious and safe, elicit lasting immunity, and cover a range of SARS-CoV-2 variants. Recombinant viral receptor-binding domains (RBDs) are safe vaccine candidates but often have limited efficacy due to the lack of virus-like immunogen display pattern. Here we have developed a novel virus-like nanoparticle (VLP) vaccine that displays 120 copies of SARS-CoV-2 RBD on its surface. This VLP-RBD vaccine mimics virus-based vaccines in immunogen display, which boosts its efficacy, while maintaining the safety of protein-based subunit vaccines. Compared to the RBD vaccine, the VLP-RBD vaccine induced five times more neutralizing antibodies in mice that efficiently blocked SARS-CoV-2 from attaching to its host receptor and potently neutralized the cell entry of variant SARS-CoV-2 strains, SARS-CoV-1, and SARS-CoV-1-related bat coronavirus. These neutralizing immune responses induced by the VLP-RBD vaccine did not wane during the two-month study period. Furthermore, the VLP-RBD vaccine effectively protected mice from SARS-CoV-2 challenge, dramatically reducing the development of clinical signs and pathological changes in immunized mice. The VLP-RBD vaccine provides one potentially effective solution to controlling the spread of SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , COVID-19/prevention & control , Immunogenicity, Vaccine , Nanoparticles/therapeutic use , Angiotensin-Converting Enzyme 2/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Drug Design , Female , HEK293 Cells , Humans , Lung/virology , Mice , Mice, Inbred BALB C , Protein Domains/immunology
8.
Nat Commun ; 12(1): 379, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483508

ABSTRACT

Allergic reactions occur when IgE molecules become crosslinked by antigens such as food proteins. Here we create the 'AllerScan' programmable phage display system to characterize the binding specificities of anti-allergen IgG and IgE antibodies in serum against thousands of allergenic proteins from hundreds of organisms at peptide resolution. Using AllerScan, we identify robust anti-wheat IgE reactivities in wheat allergic individuals but not in wheat-sensitized individuals. Meanwhile, a key wheat epitope in alpha purothionin elicits dominant IgE responses among allergic patients, and frequent IgG responses among sensitized and non-allergic patients. A double-blind, placebo-controlled trial shows that alpha purothionin reactivity, among others, is strongly modulated by oral immunotherapy in tolerized individuals. AllerScan may thus serve as a high-throughput platform for unbiased analysis of anti-allergen antibody specificities.


Subject(s)
Allergens/immunology , Antibodies/immunology , Epitopes/immunology , Peptide Library , Wheat Hypersensitivity/immunology , Adolescent , Adult , Allergens/genetics , Antibodies/blood , Antimicrobial Cationic Peptides/immunology , Child , Child, Preschool , Female , Humans , Hypersensitivity/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Plant Proteins/immunology , Young Adult
9.
J Neuropathol Exp Neurol ; 79(9): 986-997, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32783052

ABSTRACT

HIV-associated neurocognitive disorders (HAND) remain prevalent despite implementation of antiretroviral therapy (ART). Development of HAND is linked to mitochondrial dysfunction and oxidative stress in the brain; therefore, upregulation of antioxidant defenses is critical to curtail neuronal damage. Superoxide dismutase 2 (SOD2) is a mitochondrial antioxidant enzyme essential for maintaining cellular viability. We hypothesized that SOD2 was upregulated during retroviral infection. Using a simian immunodeficiency virus (SIV)-infected macaque model of HIV, quantitative PCR showed elevated SOD2 mRNA in cortical gray ([GM], 7.6-fold for SIV vs uninfected) and white matter ([WM], 77-fold for SIV vs uninfected) during SIV infection. Further, SOD2 immunostaining was enhanced in GM and WM from SIV-infected animals. Double immunofluorescence labeling illustrated that SOD2 primarily colocalized with astrocyte marker glial fibrillary acidic protein (GFAP) in SIV-infected animals. Interestingly, in ART-treated SIV-infected animals, brain SOD2 RNA levels were similar to uninfected animals. Additionally, using principal component analysis in a transcriptomic approach, SOD2 and GFAP expression separated SIV-infected from uninfected brain tissue. Projection of these data into a HIV dataset revealed similar expression changes, thereby validating the clinical relevance. Together, our findings suggest that novel SOD2-enhancing therapies may reduce neuroinflammation in ART-treated HIV-infected patients.


Subject(s)
AIDS Dementia Complex/enzymology , Astrocytes/enzymology , Simian Acquired Immunodeficiency Syndrome/enzymology , Superoxide Dismutase/metabolism , Animals , Anti-Retroviral Agents/pharmacology , Brain/enzymology , Macaca nemestrina , Male , Microglia/enzymology , Neurons/enzymology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus , Superoxide Dismutase/drug effects , Up-Regulation
10.
J Neurovirol ; 26(4): 511-519, 2020 08.
Article in English | MEDLINE | ID: mdl-32488843

ABSTRACT

HIV-associated neuroinflammation is primarily driven by CNS macrophages including microglia. Regulation of these immune responses, however, remains to be characterized in detail. Using the SIV/macaque model of HIV, we evaluated CNS expression of triggering receptor expressed on myeloid cells 2 (TREM2) which is constitutively expressed by microglia and contributes to cell survival, proliferation, and differentiation. Loss-of-function mutations in TREM2 are recognized risk factors for neurodegenerative diseases including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Nasu-Hakola disease (NHD); recent reports have also indicated a role for TREM2 in HIV-associated neuroinflammation. Using in situ hybridization (ISH) and qRT-PCR, TREM2 mRNA levels were found to be significantly elevated in frontal cortex of macaques with SIV encephalitis compared with uninfected controls (P = 0.02). TREM2 protein levels were also elevated as measured by ELISA of frontal cortex tissue homogenates in these animals. Previously, we characterized the expression of CSF1R (colony-stimulating factor 1 receptor) in this model; the TREM2 and CSF1R promoters both contain a PU.1 binding site. While TREM2 and CSF1R mRNA levels in the frontal cortex were highly correlated (Spearman R = 0.79, P < 0.001), protein levels were not well correlated. In SIV-infected macaques released from ART to study viral rebound, neither TREM2 nor CSF1R mRNA increased with rebound viremia. However, CSF1R protein levels remained significantly elevated unlike TREM2 (P = 0.02). This differential expression suggests that TREM2 and CSF1R play unique, distinct roles in the pathogenesis of HIV CNS disease.


Subject(s)
Encephalitis, Viral/genetics , Macaca nemestrina/immunology , Macrophages/immunology , Membrane Glycoproteins/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/immunology , Animals , Antiretroviral Therapy, Highly Active/methods , Antiviral Agents/pharmacology , Drug Administration Schedule , Encephalitis, Viral/drug therapy , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/virology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Macaca nemestrina/genetics , Macaca nemestrina/virology , Macrophages/drug effects , Macrophages/virology , Male , Membrane Glycoproteins/immunology , Microglia/drug effects , Microglia/immunology , Microglia/virology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , Trans-Activators/genetics , Trans-Activators/immunology
11.
J Neuropathol Exp Neurol ; 78(1): 78-87, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30500918

ABSTRACT

Human immunodeficiency virus-associated sensory neuropathy (HIV-SN) remains a frequent neurologic complication of HIV infection. Little is known about alterations in the peripheral nervous system during the early stages of HIV, a time when neuroprotective interventions may be most beneficial. We performed Nanostring gene expression analysis on lumbar dorsal root ganglia (DRG) from 6 simian immunodeficiency virus (SIV)-infected pigtailed macaques killed at 7 days post-inoculation and 8 uninfected controls. We found significant upregulation of many genes involved in immune signaling and activation in the DRG. Among genes related to glutamate metabolism, there was significant upregulation of glutamine synthetase (GS), while glutaminase (GLS) was downregulated. Several genes involved in the oxidative stress response also showed significant differential regulation in the DRG of 7d SIV-infected animals, with superoxide dismutase-2 (SOD2) showing the greatest median fold change compared to controls. Novel findings in the DRG were compared to corresponding brain data and further investigated at the protein level by Western blotting and immunohistochemistry. Together with our previous finding of significant epidermal nerve fiber loss at 14 days post-SIV infection, results of this study demonstrate that immune activation and altered cellular metabolism at in the DRG precede and likely contribute to early sensory nerve injury in HIV-SN.


Subject(s)
Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Animals , Macaca nemestrina , Male , Transcriptome
12.
J Neuropathol Exp Neurol ; 77(3): 199-206, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29319809

ABSTRACT

Chronic microglial activation and associated neuroinflammation are key factors in neurodegenerative diseases including HIV-associated neurocognitive disorders. Colony stimulating factor 1 receptor (CSF1R)-mediated signaling is constitutive in cells of the myeloid lineage, including microglia, promoting cell survival, proliferation, and differentiation. In amyotrophic lateral sclerosis and Alzheimers disease, CSF1R is upregulated. Inhibiting CSF1R signaling in animal models of these diseases improved disease outcomes. In our studies, CNS expression of the CSF1R ligand, colony-stimulating factor 1 (CSF1) was significantly increased in a SIV/macaque model of HIV CNS disease. Using a Nanostring nCounter immune panel, we found CSF1 overexpression was strongly correlated with upregulation of microglial genes involved in antiviral and oxidative stress responses. Using in situ hybridization, we found that CSF1R mRNA was only present in Iba-1 positive microglia. By ELISA and immunostaining with digital image analysis, SIV-infected macaques had significantly higher CSF1R levels in frontal cortex than uninfected macaques (p = 0.018 and p = 0.02, respectively). SIV-infected macaques treated with suppressive ART also had persistently elevated CSF1R similar to untreated SIV-infected macaques. Coordinate upregulation of CSF1 and CSF1R expression implicates this signaling pathway in progressive HIV CNS disease.


Subject(s)
Brain/pathology , Central Nervous System Diseases , Gene Expression Regulation, Viral/physiology , Microglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Simian Acquired Immunodeficiency Syndrome/complications , Animals , Brain/metabolism , Brain/virology , Central Nervous System Diseases/complications , Central Nervous System Diseases/pathology , Central Nervous System Diseases/virology , Disease Models, Animal , Interleukins/genetics , Interleukins/metabolism , Macaca nemestrina , Male , Oxidative Stress , RNA, Messenger/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
13.
Mycorrhiza ; 25(8): 611-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25771863

ABSTRACT

The Orchid flora of Madagascar is one of the most diverse with nearly 1000 orchid taxa, of which about 90% are endemic to this biodiversity hotspot. The Itremo Massif in the Central Highlands of Madagascar with a Highland Subtropical climate range encompasses montane grassland, igneous and metamorphic rock outcrops, and gallery and tapia forests. Our study focused on identifying culturable mycorrhizae from epiphytic, lithophytic, and terrestrial orchid taxa to understand their diversity and density in a spatial matrix that is within the protected areas. We have collected both juvenile and mature roots from 41 orchid taxa for isolating their orchid mycorrhizal fungi (OMF), and to culture, identify, and store in liquid nitrogen for future studies. Twelve operational taxonomic units (OTUs), of three known orchid mycorrhizal genera, were recognized by analysis of internal transcribed spacer (ITS) sequences of 85 isolates, and, by comparing with GenBank database entries, each OTU was shown to have closely related fungi that were also found as orchid associates. Orchid and fungal diversity were greater in gallery forests and open grasslands, which is very significant for future studies and orchid conservation. As far as we know, this is the first ever report of detailed identification of mycorrhizal fungi from Madagascar. This study will help start to develop a programme for identifying fungal symbionts from this unique biodiversity hotspot, which is undergoing rapid ecosystem damage and species loss. The diversity of culturable fungal associates, their density, and distribution within the Itremo orchid hotspot areas will be discussed.


Subject(s)
Mycorrhizae/classification , Mycorrhizae/isolation & purification , Orchidaceae/microbiology , Biodiversity , DNA, Fungal/analysis , DNA, Fungal/isolation & purification , Grassland , Madagascar , Molecular Sequence Data , Mycorrhizae/genetics , Phylogeny , Plant Roots/microbiology , Rhizoctonia/genetics , Rhizoctonia/growth & development , Rhizoctonia/isolation & purification , Soil Microbiology , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL
...