[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (74)

Search Parameters:
Keywords = dimorphic fungi

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 3504 KiB  
Article
On the Origins of Symbiotic Fungi in Carmine Cochineals and Their Function in the Digestion of Plant Polysaccharides
by Pilar González-Román, Diana Hernández-Oaxaca, Rafael Bustamante-Brito, Marco A. Rogel and Esperanza Martínez-Romero
Insects 2024, 15(10), 783; https://doi.org/10.3390/insects15100783 - 9 Oct 2024
Viewed by 409
Abstract
The cochineal insect Dactylopius coccus Costa (Hemiptera) has cultural and economic value because it produces carminic acid that is used commercially. In this study, distinct fungi were cultured from dissected tissue and identified as Penicillium, Coniochaeta, Arthrinium, Cladosporium, Microascus [...] Read more.
The cochineal insect Dactylopius coccus Costa (Hemiptera) has cultural and economic value because it produces carminic acid that is used commercially. In this study, distinct fungi were cultured from dissected tissue and identified as Penicillium, Coniochaeta, Arthrinium, Cladosporium, Microascus, Aspergillus, and Periconia. Fungi were microscopically observed inside cochineals in the gut, fat body, and ovaries. Since cochineals spend their lives attached to cactus leaves and use the sap as feed, they can obtain fungi from cacti plants. Indeed, we obtained Penicillium, Aspergillus, and Cladosporium fungi from cacti that were identical to those inside cochineals, supporting their plant origin. Fungi could be responsible for the degrading activities in the insect guts, since cellulase, pectinase, and amylase enzymatic activities in insect guts decreased in fungicide-treated cochineals. Our findings set the basis for the further study of the interactions between insects, fungi, and their host plants. Full article
(This article belongs to the Special Issue Ecologically Important Symbioses in Insects)
Show Figures

Figure 1

Figure 1
<p>Fungal isolates culture plates belonging to (<b>I</b>) <span class="html-italic">Periconia</span> sp., (<b>II</b>) <span class="html-italic">Aspergillius hiratsukae</span>, (<b>III</b>) <span class="html-italic">Arthrinium gutiae</span>, (<b>IV</b>) <span class="html-italic">Penicillium brevicompactum</span>, (<b>V</b>) <span class="html-italic">Penicillium olsonii</span>, (<b>VI</b>) <span class="html-italic">Cladosporium cladosporioides</span>, (<b>VII</b>) <span class="html-italic">Coniochaeta</span> sp., and (<b>VII</b>) <span class="html-italic">Microascus verrucosus</span>.</p>
Full article ">Figure 2
<p>Micrographs of <span class="html-italic">Dactylopius coccus</span> tissues: gut, fat body, and ovary, stained with lactophenol blue, calcofluor white, nigrosin, and DAPI, showing hyphae-like structures (indicated with arrows).</p>
Full article ">Figure 3
<p>(<b>A</b>) ERIC-PCR patterns of <span class="html-italic">Penicillium</span> isolates from <span class="html-italic">Dactylopius coccus</span> (PbD and PoD) and <span class="html-italic">Opuntia</span> (PbO). (1) 1 Kb marker, (2) <span class="html-italic">P. brevicompactum</span> PDAC0H1, (3) <span class="html-italic">P. brevicompactum</span> PDAG-1H1, (4) <span class="html-italic">P. brevicompactum</span> PDAG0H2 from <span class="html-italic">D. coccus</span>, (5) <span class="html-italic">Penicillium olsonni</span> PDAO-1H1 from <span class="html-italic">D. coccus</span>, (6) <span class="html-italic">P. brevicompactum</span> 31N, (7) <span class="html-italic">P. brevicompactum</span> 9N from <span class="html-italic">Opuntia</span> plants. (<b>B</b>) Molecular phylogeny of fungal ITS1-2 with Maximum Likelihood and Kimura 2-parameter model with 1,000 bootstraps.</p>
Full article ">Figure 4
<p>Thin-layer chromatography of sugars produced by the gut extract after 5 min (SA) and 3 h (SB) incubation with starch, after 5 min (CA) and 3 h (CB) incubation with cellulose, and after 5 min (PA) and 3 h (PB) incubation with pectin, compared to gut extract without substrate (C). Glucose (G) and galacturonic acid (GA) were used as standards.</p>
Full article ">Figure 5
<p>Enzymatic activities of cellulase, pectinase, and amylase before (BT) and after (AT) treatment with fludioxonil fungicide according to DNS assay. Measurements provided as g/L of reducing sugars.</p>
Full article ">Figure 6
<p><span class="html-italic">Coniochaeta</span> dimorphic morphology (<b>A</b>) yeast-like (<b>B</b>) filamentous form.</p>
Full article ">
34 pages, 3319 KiB  
Review
Diagnosis of Human Endemic Mycoses Caused by Thermally Dimorphic Fungi: From Classical to Molecular Methods
by Joaquina María García-Martín, Antonio Muro and Pedro Fernández-Soto
J. Fungi 2024, 10(9), 637; https://doi.org/10.3390/jof10090637 - 6 Sep 2024
Viewed by 860
Abstract
Human endemic mycoses are potentially fatal diseases caused by a diverse group of fungi that can alter their morphology in response to an increase in temperature. These thermally dimorphic fungi affect both healthy and immunocompromised hosts, causing a substantial health and economic burden. [...] Read more.
Human endemic mycoses are potentially fatal diseases caused by a diverse group of fungi that can alter their morphology in response to an increase in temperature. These thermally dimorphic fungi affect both healthy and immunocompromised hosts, causing a substantial health and economic burden. Despite this, the diagnosis of endemic mycoses is still a formidable challenge for several reasons, including similar symptomatology, limited utility of classical diagnostic methods, inaccessibility to reliable molecular approaches in most endemic areas, and a lack of clinical suspicion out of these regions. This review summarizes essential knowledge on thermally dimorphic fungi and the life-threatening diseases they cause. The principle, advantages and limitations of the methods traditionally used for their diagnosis are also described, along with the application status and future directions for the development of alternative diagnostic strategies, which could help to reduce the disease burden in endemic areas. Full article
(This article belongs to the Special Issue Diagnosis of Human Pathogenic Fungi)
Show Figures

Figure 1

Figure 1
<p>Distribution of relevant human pathogenic species across fungi, based on the phylogenomic tree published by Li and colleagues [<a href="#B4-jof-10-00637" class="html-bibr">4</a>] and the multigene phylogeny of the order Onygenales published by Kandemir and collaborators [<a href="#B5-jof-10-00637" class="html-bibr">5</a>]. In the genome-scale tree of fungi, derived from a concatenated data matrix (290 genes), the nine lineages currently accepted are named and shaded in different colors. Terminals of the tree are labeled using order-level taxonomic names, except for Saccharomycotina, for which informal and family-level names of the major clades are used. Orders including pathogenic species appear in bold and are colored, with those three comprising thermally dimorphic fungi of clinical interest marked with an asterisk. The phylogeny of Onygenales (lower right corner of the figure) was obtained by combining eight loci. Only those clades comprising human pathogens are formally named. A. <span class="html-italic">Trichosporon</span> spp. B. <span class="html-italic">Cryptococcus</span> spp. C. <span class="html-italic">Exobasidium</span> spp. D. <span class="html-italic">Ustilago</span> spp. E. <span class="html-italic">Mucor</span> spp. F. <span class="html-italic">Candida</span> spp. G. <span class="html-italic">Fusarium</span> spp. H. <span class="html-italic">Sporothrix</span> spp. I. <span class="html-italic">Fonsecaea</span> spp. J. <span class="html-italic">Talaromyces marneffei</span>. K. <span class="html-italic">Aspergillus</span> spp. L. <span class="html-italic">Coccidioides</span> spp. M. <span class="html-italic">Blastomyces</span> spp. N. <span class="html-italic">Emergomyces</span> spp. O. <span class="html-italic">Histoplasma</span> spp. P. <span class="html-italic">Paracoccidioides</span> spp.</p>
Full article ">Figure 2
<p>Geographic distribution of endemic mycoses based on published data [<a href="#B18-jof-10-00637" class="html-bibr">18</a>,<a href="#B39-jof-10-00637" class="html-bibr">39</a>,<a href="#B51-jof-10-00637" class="html-bibr">51</a>,<a href="#B52-jof-10-00637" class="html-bibr">52</a>,<a href="#B53-jof-10-00637" class="html-bibr">53</a>,<a href="#B54-jof-10-00637" class="html-bibr">54</a>]. (<b>A</b>) Blastomycosis; (<b>B</b>) Coccidioidomycosis; (<b>C</b>) Emergomycosis; (<b>D</b>) Histoplasmosis; (<b>E</b>) Paracoccidioidomycosis; (<b>F</b>) Sporotrichosis; and (<b>G</b>) Talaromycosis. Except for the maps of coccidioidomycosis (<b>B</b>) and emergomycosis (<b>C</b>), in which different colors represent different species, color shades represent different levels of incidence, with darker tones corresponding to the highest incidence.</p>
Full article ">Figure 3
<p>Binary heatmap showing the existence/absence of antigen and antibody detection methods (x-axis) for the diagnosis of different endemic mycoses (y-axis). Shaded cells represent the existence of a test for a given mycosis. References to studies using these methods (or citing their use) are indicated between square brackets. An asterisk indicates that at least one commercial test is available (others may exist). For more details, see the review by Cáceres and colleagues [<a href="#B117-jof-10-00637" class="html-bibr">117</a>] and <a href="https://www.immy.com" target="_blank">https://www.immy.com</a> (accessed on 31 July 2024). Abbreviations: Ab = antibody; Ag = antigen; BM = blastomycosis; CF = complement fixation; CM = coccidioidomycosis; EIA = enzyme immunoassay; ELISA = enzyme-linked immunosorbent assays; EM = emergomycosis; HP = histoplasmosis; ID = immunodiffusion; LFA = lateral flow assay; LA = latex agglutination; PCM = paracoccidioidomycosis; ST = sporotrichosis; TM = talaromycosis; WB = Western blot. References in this figure: [<a href="#B76-jof-10-00637" class="html-bibr">76</a>,<a href="#B117-jof-10-00637" class="html-bibr">117</a>,<a href="#B119-jof-10-00637" class="html-bibr">119</a>,<a href="#B120-jof-10-00637" class="html-bibr">120</a>,<a href="#B121-jof-10-00637" class="html-bibr">121</a>,<a href="#B122-jof-10-00637" class="html-bibr">122</a>,<a href="#B123-jof-10-00637" class="html-bibr">123</a>,<a href="#B124-jof-10-00637" class="html-bibr">124</a>,<a href="#B125-jof-10-00637" class="html-bibr">125</a>,<a href="#B126-jof-10-00637" class="html-bibr">126</a>,<a href="#B127-jof-10-00637" class="html-bibr">127</a>,<a href="#B128-jof-10-00637" class="html-bibr">128</a>,<a href="#B129-jof-10-00637" class="html-bibr">129</a>,<a href="#B130-jof-10-00637" class="html-bibr">130</a>,<a href="#B131-jof-10-00637" class="html-bibr">131</a>,<a href="#B132-jof-10-00637" class="html-bibr">132</a>,<a href="#B133-jof-10-00637" class="html-bibr">133</a>,<a href="#B134-jof-10-00637" class="html-bibr">134</a>,<a href="#B135-jof-10-00637" class="html-bibr">135</a>,<a href="#B136-jof-10-00637" class="html-bibr">136</a>,<a href="#B137-jof-10-00637" class="html-bibr">137</a>,<a href="#B138-jof-10-00637" class="html-bibr">138</a>,<a href="#B139-jof-10-00637" class="html-bibr">139</a>,<a href="#B140-jof-10-00637" class="html-bibr">140</a>,<a href="#B141-jof-10-00637" class="html-bibr">141</a>,<a href="#B142-jof-10-00637" class="html-bibr">142</a>,<a href="#B143-jof-10-00637" class="html-bibr">143</a>,<a href="#B144-jof-10-00637" class="html-bibr">144</a>,<a href="#B145-jof-10-00637" class="html-bibr">145</a>,<a href="#B146-jof-10-00637" class="html-bibr">146</a>,<a href="#B147-jof-10-00637" class="html-bibr">147</a>,<a href="#B148-jof-10-00637" class="html-bibr">148</a>,<a href="#B149-jof-10-00637" class="html-bibr">149</a>,<a href="#B150-jof-10-00637" class="html-bibr">150</a>,<a href="#B151-jof-10-00637" class="html-bibr">151</a>,<a href="#B152-jof-10-00637" class="html-bibr">152</a>,<a href="#B153-jof-10-00637" class="html-bibr">153</a>,<a href="#B154-jof-10-00637" class="html-bibr">154</a>].</p>
Full article ">Figure 4
<p>Timeline of PCR-based assays developed for the detection of different thermally dimorphic fungi. Target organisms (represented in different colors: dark blue—<span class="html-italic">Blastomyces</span> spp.; light blue—<span class="html-italic">Coccidioides</span> spp.; brown—<span class="html-italic">Emergomyces</span> spp.; purple—<span class="html-italic">Histoplasma</span> spp.; red—<span class="html-italic">Paracoccidioide</span>s spp.; yellow—<span class="html-italic">Talaromyces</span> spp.; green—<span class="html-italic">Sporothrix</span> spp.), type of assay (PCR, nested PCR, qPCR, or multiplex qPCR), and amplified gene(s) are indicated below each reference. For easy presentation, this graph shows only a selection of studies published in the last two decades. Most studies were based on clinical samples; otherwise, it is indicated between brackets after the gene name. References in this figure: [<a href="#B78-jof-10-00637" class="html-bibr">78</a>,<a href="#B185-jof-10-00637" class="html-bibr">185</a>,<a href="#B186-jof-10-00637" class="html-bibr">186</a>,<a href="#B187-jof-10-00637" class="html-bibr">187</a>,<a href="#B188-jof-10-00637" class="html-bibr">188</a>,<a href="#B193-jof-10-00637" class="html-bibr">193</a>,<a href="#B194-jof-10-00637" class="html-bibr">194</a>,<a href="#B195-jof-10-00637" class="html-bibr">195</a>,<a href="#B196-jof-10-00637" class="html-bibr">196</a>,<a href="#B197-jof-10-00637" class="html-bibr">197</a>,<a href="#B198-jof-10-00637" class="html-bibr">198</a>,<a href="#B199-jof-10-00637" class="html-bibr">199</a>,<a href="#B200-jof-10-00637" class="html-bibr">200</a>,<a href="#B201-jof-10-00637" class="html-bibr">201</a>,<a href="#B202-jof-10-00637" class="html-bibr">202</a>,<a href="#B203-jof-10-00637" class="html-bibr">203</a>,<a href="#B204-jof-10-00637" class="html-bibr">204</a>,<a href="#B205-jof-10-00637" class="html-bibr">205</a>,<a href="#B206-jof-10-00637" class="html-bibr">206</a>,<a href="#B207-jof-10-00637" class="html-bibr">207</a>,<a href="#B208-jof-10-00637" class="html-bibr">208</a>,<a href="#B209-jof-10-00637" class="html-bibr">209</a>,<a href="#B210-jof-10-00637" class="html-bibr">210</a>,<a href="#B211-jof-10-00637" class="html-bibr">211</a>].</p>
Full article ">Figure 5
<p>Timeline of isothermal amplification assays developed for the detection of different thermally dimorphic fungi. Target organisms (represented in different colors), type of assay (LAMP or RCA), and amplified genes are indicated for each reference. * No information available. References in this figure: [<a href="#B217-jof-10-00637" class="html-bibr">217</a>,<a href="#B218-jof-10-00637" class="html-bibr">218</a>,<a href="#B219-jof-10-00637" class="html-bibr">219</a>,<a href="#B220-jof-10-00637" class="html-bibr">220</a>,<a href="#B221-jof-10-00637" class="html-bibr">221</a>,<a href="#B222-jof-10-00637" class="html-bibr">222</a>,<a href="#B223-jof-10-00637" class="html-bibr">223</a>,<a href="#B224-jof-10-00637" class="html-bibr">224</a>].</p>
Full article ">
27 pages, 3962 KiB  
Article
Epiphytic Yeasts from South Romania for Preventing Food Microbial Contamination
by Viorica Maria Corbu, Andreea Ștefania Dumbravă, Irina Gheorghe-Barbu and Ortansa Csutak
Life 2024, 14(9), 1087; https://doi.org/10.3390/life14091087 - 29 Aug 2024
Viewed by 554
Abstract
Epiphytic yeasts represent an important source for the development of novel strategies aiming to combat food microbial contamination. The present study deals with the characterization of nine yeast strains belonging to Starmerella, Candida, Metschinikowia, Lachancea, Kodamaea and Pichia genera, [...] Read more.
Epiphytic yeasts represent an important source for the development of novel strategies aiming to combat food microbial contamination. The present study deals with the characterization of nine yeast strains belonging to Starmerella, Candida, Metschinikowia, Lachancea, Kodamaea and Pichia genera, isolated from the surface of plants from the Botanical Garden “Dimitrie Brandza” (Bucharest, Romania) for use as antimicrobial and probiotic agents. The tests involved the determination of the safe status, cell growth under stress conditions, and activity against pathogenic Candida and bacteria strains, respectively, as well as phytopathogenic filamentous fungi and lipolytic activity. None of the nine strains showed all the characteristics for virulence and pathogenicity, with the rare positive results being explained rather by their adaptability to the habitats of origin. The strains Lachancea thermotolerans CMGB-ST12 and Kodamaea ohmeri CMGB-ST19 grew at 37 °C; Metschnikowia reukaufii CMGB-ST21.2, M. reukaufii CMGB-ST.8.1 and M. reukaufii CMGB ST10 grew in the presence of 10% NaCl, while L. thermotolerans CMGB-ST12 and K. ohmeri CMGB-ST19 tolerated both acidic and alkaline pH values well (3.0 to 12.0). The studied yeast strains showed good antimicrobial activity against Candida krusei, Candida albicans and Gram-negative bacterial strains, with K. ohmeri CMGB-ST19 and Pichia membranaefaciens CMGB-ST53 inhibiting up to 100% the development of filamentous fungi. All the strains produced lipases for tributyrin hydrolysis, the best producer being Starmerella bombi CMGB-ST1, and only Candida magnoliae CMGB-ST8.2 tested positive against other probiotic yeasts. Overall, our nine yeast strains show high potential for industrial applications, for obtaining probiotic products and for preventing the development of a wide range of microbial food contaminants. Full article
(This article belongs to the Special Issue Food Microbiological Contamination)
Show Figures

Figure 1

Figure 1
<p>Phenotypic analysis of soluble virulence factors produced by different yeast strains ((<b>A</b>)—caseinases; (<b>B</b>)—siderophore-like compounds; (<b>C</b>)—amylolytic enzymes) (yellow arrow indicates positive result obtained for <span class="html-italic">M. reukaufii</span> CMGB-ST21.1 strain).</p>
Full article ">Figure 2
<p><span class="html-italic">P. membranaefaciens</span> CMGB-ST53 cell appearance (40×) after 24 h of incubation on YPG medium (<b>A</b>), respectively, after 3 h in the presence of fetal bovine serum (<b>B</b>).</p>
Full article ">Figure 3
<p>Growth of yeast strains after 24 h of cultivation at different temperature (°C) values ((<b>A</b>)—<span class="html-italic">M. reukaufii</span> strains; (<b>B</b>)—other yeast strains) (Tukey test, <span class="html-italic">p</span> &lt; 0.05)).</p>
Full article ">Figure 4
<p>Growth of yeast strains after 24 h of cultivation at different NaCl (%) concentrations ((<b>A</b>)—<span class="html-italic">M. reukaufii</span> strains; (<b>B</b>)—other yeast strains) (Tukey test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Growth of yeast strains after 24 h of cultivation in presence of different pH values ((<b>A</b>)—<span class="html-italic">M. reukaufii</span> strains; (<b>B</b>)—other yeast strains) (Tukey test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>The interaction between the yeast strains and <span class="html-italic">A. flavus</span> T11—(<b>A1</b>–<b>A3</b>); <span class="html-italic">A. ochraceus</span>—(<b>B1</b>–<b>B3</b>); <span class="html-italic">B. cinereea</span>—(<b>C1</b>–<b>C3</b>) (1;2—yeasts-fungi interaction; 3—control).</p>
Full article ">Figure 7
<p>Determination of the inhibition percentage of mycelial growth in the case of strains of filamentous fungi belonging to the genus <span class="html-italic">Aspergillus</span> after 5 days of incubation (<b>A</b>) and 8 days of incubation (<b>B</b>) (Tukey test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
14 pages, 4955 KiB  
Article
Fruiting Body Heterogeneity, Dimorphism and Haustorium-like Structure of Naematelia aurantialba (Jin Er Mushroom)
by Ying Yang and Caihong Dong
J. Fungi 2024, 10(8), 557; https://doi.org/10.3390/jof10080557 - 7 Aug 2024
Viewed by 581
Abstract
Mushroom Jin Er has attracted widespread attention in Asia over the past two decades due to its medicinal properties and nutritional values. In the present study, Jin Er basidiocarps were often found to be surrounded by Stereum hirsutum fruiting bodies in their natural [...] Read more.
Mushroom Jin Er has attracted widespread attention in Asia over the past two decades due to its medicinal properties and nutritional values. In the present study, Jin Er basidiocarps were often found to be surrounded by Stereum hirsutum fruiting bodies in their natural habitat and occasionally in artificial cultivation. The observation of two different kinds of mycelia within the hymenium and analyses of ITS sequences confirmed that Jin Er basidiocarps were composed of two fungal species, Naematelia aurantialba and S. hirsutum. This heterogeneity of Jin Er fruiting bodies is indeed distinct from the homogeneous hypha of Tremella fuciformis found in Yin Er mushroom, although its development also requires the presence of another fungus Annulohypoxylon stygium. Basidiospores can germinate on the surface of basidiocarps and produce mycelia. However, basidiospores in PDA medium can only bud into yeast-like conidia. The yeast-like conidia of N. aurantialba can transform into pseudohyphae with a change in temperature from 20 °C to 28 °C or switch into filamentous cells on an induction medium (IDM) at 20 °C, 25 °C and 28 °C. This dimorphic was reported for the first time in N. aurantialba. Haustorium-like structures were abundantly observed both within the hymenium and in the aerial mycelia cultured on the IDM. The developmental process was documented firstly in this study, involving the formation of protuberances with basal clamp connections, elongation at the protuberances, branch production, and eventual maturation. However, further observation is required to determine whether the haustorium-like structures can penetrate S. hirsutum hyphae. These findings are expected to provide valuable insights into the relationship and interaction between these two fungi, thereby advancing the cultivation of fruiting bodies. Full article
Show Figures

Figure 1

Figure 1
<p>Jin Er fruiting bodies are composed of two fungal species. (<b>A</b>) Wild Jin Er fruiting bodies. <span class="html-italic">S. hirsutum</span> fruiting bodies adjacent to Jin Er are circled in red; (<b>B</b>) artificially cultivated Jin Er to scale; (<b>C</b>) artificially cultivated Jin Er and <span class="html-italic">S. hirsutum</span> on the growing bag (circled in red); (<b>D</b>) artificially cultivated Jin Er fruiting bodies; (<b>E</b>) electrophoretic bands of ITS amplification of DNA from different locations of Jin Er. (<b>F</b>) Phylogenetic tree based on the ITS sequences of <span class="html-italic">N. aurantialba</span> and related species; (<b>G</b>) phylogenetic tree based on the ITS sequences of <span class="html-italic">S. hirsutum</span> and related species. Sequences in red are from this study. Red scale bar = 1000 μm, white scale bar = 1 cm. M: marker 2 kb; NA: <span class="html-italic">N. aurantialba</span> basidiospores; SH: <span class="html-italic">S. hirsutum</span> pure mycelia; FB1, FB2 and FB3: top, middle, and bottom of Jin Er fruiting body circled in red; CK: negative control. FB2 (top): upper band of sample FB2 in (<b>E</b>); FB2 (bottom): lower band of sample FB2 in (<b>E</b>). Superior characters * and T: strain number and type species, respectively.</p>
Full article ">Figure 2
<p>Microscopic structure of Jin Er hymenium. (<b>A</b>) probasidia shown by red arrow; (<b>B</b>,<b>C</b>) mature basidia shown by red arrows; (<b>D</b>) wpibasidia shown by red arrow; (<b>E</b>): basidiospores; (<b>F</b>) conidia are indicated by red arrows. Scale bar: 10 μm.</p>
Full article ">Figure 3
<p>The differences in hyphal morphology between <span class="html-italic">Naematelia aurantialba</span> and <span class="html-italic">Stereum hirsutum</span>. (<b>A</b>) <span class="html-italic">N. aurantialba</span> and <span class="html-italic">S. hirsutum</span> hyphae in hymenium shown by white and red arrow, respectively; (<b>B</b>,<b>C</b>) Clamp connections on <span class="html-italic">N. aurantialba</span> hyphae shown by white arrows; (<b>D</b>–<b>F</b>) Clamp connections on <span class="html-italic">S. hirsutum</span> hyphae shown by red arrows. Scale bars: 10 μm.</p>
Full article ">Figure 4
<p>Germ tubes and hyphae generated from basidiospores on the surface of Jin Er. (<b>A</b>) Fruiting body with a clean surface; (<b>B</b>) fruiting body appearing yellow and white, intermingled; (<b>C</b>) surface of basidiocarps covered with white filaments; (<b>D</b>) surface of basidiocarps under a dissecting microscope; (<b>E</b>) germ tubes formed by basidiospores, shown by red arrows; (<b>F</b>) hyphae covering the surface of basidiocarps. Scale bar: 20 μm.</p>
Full article ">Figure 5
<p>Yeast-like blastoconidia formed by budding reproduction from basidiospores of <span class="html-italic">Naematelia aurantialba</span>. (<b>A</b>,<b>B</b>) Bud formation; (<b>C</b>,<b>D</b>) abscission of buds. Scale bar: 10 μm.</p>
Full article ">Figure 6
<p>Dimorphic transition between yeast-like conidia and pseudohyphae or hyphae and conidia of <span class="html-italic">Naematelia aurantialba</span> produced by hyphae. (<b>A</b>) Conidia in bud; (<b>B</b>) pseudohyphae transformed from yeast-like conidia on PDA at 28 °C; (<b>C</b>) filamentous cells formed by yeast-like conidia on IDM at 25 °C; (<b>D</b>–<b>F</b>) conidia formed by filamentous cells shown by red arrows. Scale bar: 10 μm.</p>
Full article ">Figure 7
<p>The morphology and developmental process of haustorium-like structures in Jin Er hymenium and mycelia grown from yeast-like conidia. (<b>A</b>–<b>C</b>) Haustorium-like structures on mycelia in Jin Er hymenium; (<b>D</b>–<b>F</b>) haustorium-like structures on mycelia grown from yeast-like conidia on IDM, as observed under an optical microscope; (<b>G</b>–<b>I</b>) haustorium-like structures on mycelia grown from yeast-like conidia on IDM using SEM. Red arrows show haustorium-like structures. Scale bar: 10 μm.</p>
Full article ">
8 pages, 778 KiB  
Brief Report
Coccidioidomycosis in Immunocompromised at a Non-Endemic Referral Center in Mexico
by Carla M. Román-Montes, Lisset Seoane-Hernández, Rommel Flores-Miranda, Andrea Carolina Tello-Mercado, Andrea Rangel-Cordero, Rosa Areli Martínez-Gamboa, José Sifuentes-Osornio, Alfredo Ponce-de-León and Fernanda González-Lara
J. Fungi 2024, 10(6), 429; https://doi.org/10.3390/jof10060429 - 18 Jun 2024
Viewed by 829
Abstract
The incidence and distribution of coccidioidomycosis are increasing. Information scarcity is evident in Mexico, particularly in non-endemic zones and specific populations. We compared the treatment and outcomes for patients with isolated pulmonary infections and those with disseminated coccidioidomycosis, including mortality rates within six [...] Read more.
The incidence and distribution of coccidioidomycosis are increasing. Information scarcity is evident in Mexico, particularly in non-endemic zones and specific populations. We compared the treatment and outcomes for patients with isolated pulmonary infections and those with disseminated coccidioidomycosis, including mortality rates within six weeks of diagnosis. Of 31 CM cases, 71% were male and 55% were disseminated. For 42% of patients, there was no evidence of having lived in or visited an endemic region. All patients had at least one comorbidity, and 58% had pharmacologic immunosuppressants. The general mortality rate was 30%; without differences between disseminated and localized disease. In our research, we describe a CM with a high frequency of disseminated disease without specific risk factors and non-significant mortality. Exposure to endemic regions was not found in a considerable number of subjects. We consider diverse reasons for why this may be, such as climate change or migration. Full article
(This article belongs to the Special Issue Dimorphic Fungal Pathogen Coccidioides and Coccidioidomycosis)
Show Figures

Figure 1

Figure 1
<p>This map of Mexico and the Southwestern United States shows where infections are supposed to have been acquired. It is color-coded to differentiate between known or not known to be endemic regions [<a href="#B14-jof-10-00429" class="html-bibr">14</a>].</p>
Full article ">
12 pages, 251 KiB  
Review
Histoplasmosis in Taiwan: Case Summary and Literature Review
by Jui-Chi Hsu, Po-Hsun Chang, Chien-Hsiang Tai and Yi-Chun Chen
Life 2024, 14(6), 738; https://doi.org/10.3390/life14060738 - 7 Jun 2024
Viewed by 925
Abstract
Histoplasmosis is a global infection caused by the thermally dimorphic fungus, Histoplasma capsulatum complex. It is endemic in the United States, as well as in Central and South America. In Taiwan, histoplasmosis is rare, with the first reported case not occurring until 1977. [...] Read more.
Histoplasmosis is a global infection caused by the thermally dimorphic fungus, Histoplasma capsulatum complex. It is endemic in the United States, as well as in Central and South America. In Taiwan, histoplasmosis is rare, with the first reported case not occurring until 1977. We summarized a total of 17 cases reported in Taiwan over the past 40 years and provided detailed descriptions for four probable indigenous cases. Due to the lack of rapid diagnostic tools and clinical suspicion, histoplasmosis may be underdiagnosed in Taiwan. We recognize that a limitation of our review is the lack of data on the environmental surveillance for H. capsulatum complex in Taiwan. Conducting a further phylogenetic analysis on both environmental and clinical isolates would provide valuable evidence for the region. Full article
(This article belongs to the Special Issue Fungal Disease Epidemiology: Current State and Future Prospects)
15 pages, 563 KiB  
Review
The Role of Olorofim in the Treatment of Filamentous Fungal Infections: A Review of In Vitro and In Vivo Studies
by Aliosha Feuss, Marie-Elisabeth Bougnoux and Eric Dannaoui
J. Fungi 2024, 10(5), 345; https://doi.org/10.3390/jof10050345 - 10 May 2024
Viewed by 1588
Abstract
Invasive fungal infections have recently been recognized by the WHO as a major health, epidemiological, and economic issue. Their high mortality rates and the emergence of drug resistance have driven the development of new molecules, including olorofim, an antifungal belonging to a new [...] Read more.
Invasive fungal infections have recently been recognized by the WHO as a major health, epidemiological, and economic issue. Their high mortality rates and the emergence of drug resistance have driven the development of new molecules, including olorofim, an antifungal belonging to a new family of compounds, the orotomides. A review was conducted on the PubMed database and the ClinicalTrials.gov website to summarize the microbiological profile of olorofim and its role in the treatment of filamentous fungal infections. Twenty-four articles were included from the search and divided into two groups: an “in vitro” group focusing on minimum inhibitory concentration (MIC) results for various fungi and an “in vivo” group evaluating the pharmacokinetics (PK), pharmacodynamics (PD), efficacy, and tolerability of olorofim in animal models of fungal infection and in humans. Olorofim demonstrated in vitro and in vivo activity against numerous filamentous fungi, including azole-resistant Aspergillus fumigatus, various dermatophytes, and endemic and dimorphic fungi. in vitro results showed higher MICs for certain Fusarium species and dematiaceous fungi Alternaria alternata and Exophiala dermatitidis; further in vivo studies are needed. Published PK-PD data in humans are limited. The results of the ongoing phase III clinical trial are eagerly awaited to evaluate olorofim’s clinical impact. Full article
(This article belongs to the Special Issue Azole Resistance in Aspergillus spp., 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Flux diagram of the olorofim studies selection process for the review.</p>
Full article ">
17 pages, 5234 KiB  
Article
Redundant and Distinct Roles of Two 14-3-3 Proteins in Fusarium sacchari, Pathogen of Sugarcane Pokkah Boeng Disease
by Yuejia Chen, Ziting Yao, Lixian Zhao, Mei Yu, Baoshan Chen and Chengwu Zou
J. Fungi 2024, 10(4), 257; https://doi.org/10.3390/jof10040257 - 28 Mar 2024
Viewed by 1259
Abstract
Fusarium sacchari, a key pathogen of sugarcane, is responsible for the Pokkah boeng disease (PBD) in China. The 14-3-3 proteins have been implicated in critical developmental processes, including dimorphic transition, signal transduction, and carbon metabolism in various phytopathogenic fungi. However, their roles [...] Read more.
Fusarium sacchari, a key pathogen of sugarcane, is responsible for the Pokkah boeng disease (PBD) in China. The 14-3-3 proteins have been implicated in critical developmental processes, including dimorphic transition, signal transduction, and carbon metabolism in various phytopathogenic fungi. However, their roles are poorly understood in F. sacchari. This study focused on the characterization of two 14-3-3 protein-encoding genes, FsBmh1 and FsBmh2, within F. sacchari. Both genes were found to be expressed during the vegetative growth stage, yet FsBmh1 was repressed at the sporulation stage in vitro. To elucidate the functions of these genes, the deletion mutants ΔFsBmh1 and ΔFsBmh2 were generated. The ΔFsBmh2 exhibited more pronounced phenotypic defects, such as impaired hyphal branching, septation, conidiation, spore germination, and colony growth, compared to the ΔFsBmh1. Notably, both knockout mutants showed a reduction in virulence, with transcriptome analysis revealing changes associated with the observed phenotypes. To further investigate the functional interplay between FsBmh1 and FsBmh2, we constructed and analyzed mutants with combined deletion and silencing (ΔFsBmh/siFsBmh) as well as overexpression (O-FsBmh). The combinations of ΔFsBmh1/siFsBmh2 or ΔFsBmh2/siFsBmh1 displayed more severe phenotypes than those with single allele deletions, suggesting a functional redundancy between the two 14-3-3 proteins. Yeast two-hybrid (Y2H) assays identified 20 proteins with pivotal roles in primary metabolism or diverse biological functions, 12 of which interacted with both FsBmh1 and FsBmh2. Three proteins were specifically associated with FsBmh1, while five interacted exclusively with FsBmh2. In summary, this research provides novel insights into the roles of FsBmh1 and FsBmh2 in F. sacchari and highlights potential targets for PBD management through the modulation of FsBmh functions. Full article
(This article belongs to the Section Fungal Pathogenesis and Disease Control)
Show Figures

Figure 1

Figure 1
<p>Expression patterns of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> at different developmental stages in vivo and in vitro. (<b>A</b>) Symptom development. Sugarcane seedlings were inoculated with CNO-1. (<b>B</b>) Development of infection. Tissue sections were stained with aniline blue to trace fungal hyphae development. Samples were observed under a light microscope, scale bar = 50 µm. (<b>C</b>) Relative expression of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> during infection. (<b>D</b>) Development of hyphae and asexual spores on liquid medium. The spore suspension of CNO-1 was inoculated in PDB medium, and representative pictures were photographed at 10 h, 14 h, and 22 h by microscope, scale bar = 50 µm. (<b>E</b>) Relative expression of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> during saprophytic stages. The expression of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> transcripts was measured by quantitative real-time RT-PCR (2<sup>−ΔΔCT</sup>method) with 18S rRNA as internal reference. The transcript levels of 0 h post-inoculation were set to a value of 1.0 and indicated by the dotted line. Different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Validation of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> expression using qRT-PCR in <span class="html-italic">FsBmh</span> deletion and complementation mutants. Relative expression of <span class="html-italic">FsBmh</span> genes in <span class="html-italic">FsBmh-</span>deletion and complementation mutants were determined by qRT-PCR. The expression of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> transcripts was measured by quantitative real-time RT-PCR (2<sup>−ΔΔCT</sup> method) with 18S rRNA as internal reference. The transcript levels of WT were set to a value of 1.0 and indicated by the dotted line. Different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Phenotypes of <span class="html-italic">FsBmh</span>-deletion mutants and complementation mutants. (<b>A</b>) Hyphal and conidial morphology. Colonies were photographed on PDA plates on day 7 after inoculation. Hyphal branches were photographed on PDA plates on day 3 after inoculation using a microscope, scale bar = 50 µm. To visualize septa, hyphae were stained with CFW and examined under a fluorescent microscope, scale bar = 10 µm. To minimize the error of comparing cells of different ages, only the fourth cells from the tip of a hypha were measured. The septa were marked by yellow arrows. Spore characteristics were photographed by a differential-interference contrast microscope, scale bar = 10 µm. (<b>B</b>) Statistics of conidial yield of strains. Conidia were harvested from colonies of 7-days-old PDA plates. (<b>C</b>) Conidial germination rate in PDB at 28 °C with rotation of 150 rpm for 6 h. Different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Phenotypes of <span class="html-italic">FsBmh-</span>deletion mutants and <span class="html-italic">FsBmh-</span>silenced mutants. (<b>A</b>) Validation of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> expression using qRT-PCR in ΔFsBmh1/siFsBmh2 and ΔFsBmh2/siFsBmh1. The expression of <span class="html-italic">FsBmh1</span> and <span class="html-italic">FsBmh2</span> transcripts was measured by quantitative real-time RT-PCR (2<sup>−ΔΔCT</sup> method) with 18S rRNA as internal reference. The transcript levels of WT were set to a value of 1.0 and indicated by the dotted line. (<b>B</b>) Hyphal and conidial morphology. Colonies were photographed on PDA plates on day 7 after inoculation. Hyphal branches were photographed on PDA plates on day 3 after inoculation using a microscope, scale bar = 50 µm. To visualize septa, hyphae were stained with CFW and examined under a fluorescent microscope, scale bar = 10 µm. To minimize the error of comparing cells of different ages, only the fourth cells from the tip of a hypha were measured. The septa were marked by yellow arrows. Spores were photographed by a differential-interference contrast microscope, scale bar = 10 µm. (<b>C</b>) Statistics of conidial yield of strains. Conidia were harvested from colonies of 7-days-old PDA plates. (<b>D</b>) Conidial germination rate in PDB at 28 °C with rotation of 150 rpm for 6 h. Different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>(<b>A</b>) Macrospores were induced in carnation leaf medium. Spores were harvested on day 7 and stained by CFW and examined under a fluorescent microscope, scale bar = 10 µm. (<b>B</b>) Classification of types of spores. Micro–M = mini microspore; Micro–N = normal microspore; Macro–2 = macrospore with 2 cells; Macro–4 = macrospore with 4 cells, scale bar = 10 µm. (<b>C</b>) Distribution of different types of spores (<span class="html-italic">n</span> = 100). The images were taken using a differential-interference contrast microscope.</p>
Full article ">Figure 6
<p><span class="html-italic">FsBmh-</span>deletion mutants are defective in virulence. Each of the plants was injected with 300 µL of spore suspension at concentration of 1 × 10<sup>4</sup> conidia/mL. (<b>A</b>) Symptoms on sugarcane seedlings. Photographs were taken 21 dpi. (<b>B</b>) Quantification of disease severity. The disease severity index was determined using 100 seedlings per treatment with 3 replicates. Different letters indicate significant difference at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Distribution of DEGs (≥two-fold) in ΔFsBmh1 and ΔFsBmh2 versus WT. (<b>A</b>) Venn diagrams showing the overlapped counts of the genes up-regulated or down-regulated in both ΔFsBmh1 and ΔFsBmh2 versus WT. (<b>B</b>) KEGG pathway enrichment of genes up-regulated or down-regulated in ΔFsBmh1 and ΔFsBmh2 versus WT.</p>
Full article ">Figure 8
<p>Hierarchical clustering of DEGs among WT, ΔFsBmh1, and ΔFsBmh2. (<b>A</b>) A total of 981 DEGs that were enriched in KEGG pathways (ΔFsBmh1 and ΔFsBmh2 versus WT) were clustered into 10 clusters. Red indicates high expression, blue indicates low expression. (<b>B</b>) Gene expression values on selected clusters. The dotted line indicates the average expression level of all DEGs within the cluster and its value was set as 0. Color scale shows the level of gene expression of log10<sup>(FPKM+ 1)</sup>.</p>
Full article ">
18 pages, 366 KiB  
Review
The Ecology of Non-Candida Yeasts and Dimorphic Fungi in Cetaceans: From Pathogenicity to Environmental and Global Health Implications
by Victor Garcia-Bustos, Begoña Acosta-Hernández, Marta Dafne Cabañero-Navalón, Javier Pemán, Alba Cecilia Ruiz-Gaitán and Inmaculada Rosario Medina
J. Fungi 2024, 10(2), 111; https://doi.org/10.3390/jof10020111 - 29 Jan 2024
Cited by 1 | Viewed by 1873
Abstract
Cetaceans, which are integral to marine ecosystems, face escalating anthropogenic threats, including climate change and pollution, positioning them as critical sentinel species for ocean and human health. This review explores the neglected realm of non-Candida yeasts in cetaceans, addressing the gaps in [...] Read more.
Cetaceans, which are integral to marine ecosystems, face escalating anthropogenic threats, including climate change and pollution, positioning them as critical sentinel species for ocean and human health. This review explores the neglected realm of non-Candida yeasts in cetaceans, addressing the gaps in the understanding of their prevalence, pathogenicity, and environmental impacts. By examining identified species such as Cryptococcus spp., Paracoccidioides spp., and several dimorphic fungi, this review emphasizes global prevalence, epidemiology and ecology, pathogenicity, and potential zoonotic implications. It also discusses the fine line between yeast commensalism and pathogenicity by considering environmental influences such as pollution, climate shifts, and immune suppression. Environmental impact discussions delve into how rising ocean temperatures and pollution can modify yeast mycobiota, potentially affecting marine host health and broader ecosystem dynamics. The cetacean’s unique physiology and ecological niches are considered, highlighting potential impacts on behaviors, reproductive success, and survival rates. Identifying crucial knowledge gaps, the review calls for intensified research efforts, employing advanced molecular techniques to unravel the cetacean mycobiome. Systematic studies on yeast diversity, antifungal susceptibility, and their influence on environmental and ecosystem health are proposed, and the balance between commensal and pathogenic species emphasizes the significance of the One Health approach. In conclusion, as marine mammals face unprecedented challenges, unveiling non-Candida yeasts in cetaceans emerges as a critical endeavor with far-reaching implications for the conservation of marine ecosystems and for both animal and human public health. Full article
(This article belongs to the Special Issue Fungal Diseases in Animals, 2nd Edition)
16 pages, 5725 KiB  
Article
Volatile Fingerprint Mediates Yeast-to-Mycelial Conversion in Two Strains of Beauveria bassiana Exhibiting Varied Virulence
by Arturo Ramírez-Ordorica, José Alberto Patiño-Medina, Víctor Meza-Carmen and Lourdes Macías-Rodríguez
J. Fungi 2023, 9(12), 1135; https://doi.org/10.3390/jof9121135 - 24 Nov 2023
Cited by 1 | Viewed by 1313
Abstract
Beauveria bassiana is a dimorphic and entomopathogenic fungus with different ecological roles in nature. In pathogenic fungi, yeast-to-mycelial conversion, which is controlled by environmental factors, is required for virulence. Here, we studied the effects of different stimuli on the morphology of two B. [...] Read more.
Beauveria bassiana is a dimorphic and entomopathogenic fungus with different ecological roles in nature. In pathogenic fungi, yeast-to-mycelial conversion, which is controlled by environmental factors, is required for virulence. Here, we studied the effects of different stimuli on the morphology of two B. bassiana strains and compared the toxicities of culture filtrates. In addition, we explored the role of volatiles as quorum sensing-like signals during dimorphic transition. The killing assays in Caenorhabditis elegans (Nematoda: Rhabditidae) showed that strain AI2 isolated from a mycosed insect cadaver had higher toxicity than strain AS5 isolated from soil. Furthermore, AI2 showed earlier yeast-to-mycelial switching than AS5. However, an increase in inoculum size induced faster yeast-to-mycelium conversion in AS5 cells, suggesting a cell-density-dependent phenomenon. Gas chromatography-mass spectrometry (GC-MS) analyses showed that the fingerprint of the volatiles was strain-specific; however, during the morphological switching, an inverse relationship between the abundance of total terpenes and 3-methylbutanol was observed in both strains. Fungal exposure to 3-methylbutanol retarded the yeast-to-mycelium transition. Hence, this study provides evidence that volatile compounds are associated with critical events in the life cycle of B. bassiana. Full article
Show Figures

Figure 1

Figure 1
<p>Survival rate of <span class="html-italic">C. elegans</span> reared in the presence of the secretome of <span class="html-italic">B. bassiana.</span> The fungal strains AI2 and AS5 were grown on PDB culture medium, and the cell-free media were used to perform the assays. A total of 15 nematodes were used per experiment and incubated at 18 °C for 72 h. n = 3, with three independent repetitions. Data were statistically analyzed using the Mantel–Cox test; **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Development of <span class="html-italic">B. bassiana</span> AI2 (<b>A</b>) and AS5 (<b>B</b>) strains inoculated on MacConkey agar medium and exposed to 3-methylbutanol (3MB). The inoculum size was 10<sup>6</sup> spores. The experiment was conducted in triplicate. Data are presented as mean ± standard error. Asterisks indicate means that differed significantly at * <span class="html-italic">p</span> ˂ 0.05, ** <span class="html-italic">p</span> ˂ 0.01, *** <span class="html-italic">p</span> ˂ 0.001 based on one-way ANOVA and Fisher’s LSD test. Means within columns with different letter indicate statistically significant differences using one-way ANOVA and Tukey’s test (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 3
<p>Effect of the carbon and nitrogen sources and oxygen on the morphology of the strain AS5 of <span class="html-italic">B. bassiana</span> at 24 h of fungal growth. Conidia were germinated in PDB, YPG-2%, YPG-6% and YNB for 24 h at 28 °C (anaerobiosis) and 150 rpm (aerobiosis). (<b>A</b>) Germination kinetics under aerobiosis conditions. Representative photographs at 100× of the morphology in (<b>B</b>) aerobiosis and (<b>C</b>) anaerobiosis at 24 h. White arrowheads indicate germinated conidia and white arrows indicate ungerminated conidia. Scale bar, 20 μm. n = 3, with three independent repetitions. Data were analyzed with one-way ANOVA and Fisher post hoc test (***, <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 4
<p>Effect of pH on the morphology of the strain AS5 from <span class="html-italic">B. bassiana</span>. Conidia were germinated in PDB at pH 3, 6, and 9 for 24 h at 28 °C (anaerobiosis) and 150 rpm (aerobiosis). (<b>A</b>) Germination kinetics under aerobiosis conditions. Representative photographs at 100× of the morphology during (<b>B</b>) aerobiosis and (<b>C</b>) anaerobiosis at 24 h. White arrowheads indicate germinated conidia and white arrows indicate ungerminated conidia. Scale bar, 20 μm. Data were analyzed with one-way ANOVA and Fisher post hoc test (***, <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>Effect of inoculum size on the morphology of <span class="html-italic">B. bassiana</span>. (<b>A</b>) Spores of the AS5 and AI2 strains (100, 500, and 1000) were inoculated in Petri dishes with 30 mL MacConkey agar medium. Representative photographs of the morphology for AS5 and AI2 at 5 and 6 d of the fungal growth. AS5 was developed as yeast cells in all inoculum sizes, whereas AI2 switched to mycelium phase on day 6. (<b>B</b>) Representative photographs of the mycelial growth for both strains on day 2 with an inoculum size of 10<sup>7</sup> spores.</p>
Full article ">Figure 6
<p>Principal component analysis (PCA) plots obtained based on the data of volatiles emitted by AI2 (<b>A</b>) and AS5 (<b>B</b>) strains on MacConkey agar medium and analyzed via GC-MS at different times of the fungal growth (n = 3). PERMANOVA (α = 0.05). (<b>C</b>,<b>D</b>) Plots depicting the contribution of the compounds to PCA using the relative peak areas (%) obtained in the chromatograms from AI2 and AS5, respectively.</p>
Full article ">Figure 7
<p>Representative microscopic image recorded at 40× for hyphal bodies, conidia, and mycelium of <span class="html-italic">B. bassiana</span> AS5 at 72 h of fungal growth and during exposure to 3-methylbutanol. An inoculum size of 10<sup>6</sup> spores was inoculated on MacConkey agar culture medium. White arrowheads indicate yeast-like cells and white arrows indicate conidia.</p>
Full article ">
11 pages, 742 KiB  
Systematic Review
Emergomycosis, an Emerging Thermally Dimorphic Fungal Infection: A Systematic Review
by Kalaiselvi Vinayagamoorthy, Dinesh Reddy Gangavaram, Anna Skiada and Hariprasath Prakash
J. Fungi 2023, 9(10), 1039; https://doi.org/10.3390/jof9101039 - 23 Oct 2023
Cited by 6 | Viewed by 2286
Abstract
Emergomycosis is an endemic mycosis caused by the Emergomyces species. Infections due to this agent have been reported globally. Hence, the present systematic review on Emergomyces infections was conducted to study the disease epidemiology, underlying diseases and risk factors, causative agents, and treatment [...] Read more.
Emergomycosis is an endemic mycosis caused by the Emergomyces species. Infections due to this agent have been reported globally. Hence, the present systematic review on Emergomyces infections was conducted to study the disease epidemiology, underlying diseases and risk factors, causative agents, and treatment and outcome. The MEDLINE, Scopus, Embase, and Web of Science databases were searched systematically with appropriate keywords from January 1990 to October 2022. A total of 77 cases of emergomycosis were included in the analysis. Emergomycosis was most commonly seen in patients with human immunodeficiency virus (HIV) infection (n = 61, 79.2%) and HIV-uninfected patients with or without other comorbidities (n = 16, 20.8%). The underlying disease and risk factors significantly associated with emergomycosis in the HIV-infected patients were CD4+ T-cell counts less than 100 cells/mm3 (n = 55, 90.2%), anaemia (n = 30, 49.2%), and thrombocytopenia (n = 17, 27.9%), whereas in the HIV-uninfected patients, treatment with immunosuppressive drugs (n = 10, 62.5%), renal disease (n = 8, 50%), transplant recipients (n = 6, 37.5%), and diabetes mellitus (n = 4, 25%) were the significant risk factors associated with emergomycosis. Emergomyces africanus (n = 55, 71.4%) is the most common causative agent, followed by E. pasteurianus (n = 9, 11.7%) and E. canadensis (n = 5, 6.5%). E. africanus was most often isolated from HIV-infected patients (n = 54, 98.2%), whereas E. pasteurianus was most common in HIV-uninfected patients (n = 5, 55.6%). The all-cause mortality rate of the total cohort is 42.9%. No significant variation in the mortality rate is observed between the HIV-infected patients (n = 28, 36.4%) and the HIV-uninfected patients (n = 5, 6.5%). In conclusion, with an increase in the immunosuppressed population across the globe in addition to HIV infection, the case burden of emergomycosis may increase in the future. Hence, clinicians and mycologists should be vigilant and clinically suspicious of emergomycosis, which helps in early diagnosis and initiation of antifungal treatment to prevent disease mortality. Full article
(This article belongs to the Section Fungal Pathogenesis and Disease Control)
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart depicting the study design and process.</p>
Full article ">
12 pages, 823 KiB  
Review
Closing the Gap in Proteomic Identification of Histoplasma capsulatum: A Case Report and Review of Literature
by Terenzio Cosio, Roberta Gaziano, Carla Fontana, Enrico Salvatore Pistoia, Rosalba Petruccelli, Marco Favaro, Francesca Pica, Silvia Minelli, Maria Cristina Bossa, Anna Altieri, Domenico Ombres, Nikkia Zarabian and Cartesio D’Agostini
J. Fungi 2023, 9(10), 1019; https://doi.org/10.3390/jof9101019 - 15 Oct 2023
Cited by 1 | Viewed by 2960
Abstract
Histoplasmosis is a globally distributed systemic infection caused by the dimorphic fungus Histoplasma capsulatum (H. capsulatum). This fungus can cause a wide spectrum of clinical manifestations, and the diagnosis of progressive disseminated histoplasmosis is often a challenge for clinicians. Although microscopy [...] Read more.
Histoplasmosis is a globally distributed systemic infection caused by the dimorphic fungus Histoplasma capsulatum (H. capsulatum). This fungus can cause a wide spectrum of clinical manifestations, and the diagnosis of progressive disseminated histoplasmosis is often a challenge for clinicians. Although microscopy and culture remain the gold standard diagnostic tests for Histoplasma identification, matrix-assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF MS) has emerged as a method of microbial identification suitable for the confirmation of dimorphic fungi. However, to our knowledge, there are no entries for H. capsulatum spectra in most commercial databases. In this review, we describe the case of disseminated histoplasmosis in a patient living with HIV admitted to our university hospital that we failed to identify by the MALDI-TOF method due to the limited reference spectrum of the instrument database. Furthermore, we highlight the utility of molecular approaches, such as conventional polymerase chain reaction (PCR) and DNA sequencing, as alternative confirmatory tests to MALDI-TOF technology for identifying H. capsulatum from positive cultures. An overview of current evidence and limitations of MALDI-TOF-based characterization of H. capsulatum is also presented. Full article
(This article belongs to the Special Issue Histoplasma and Histoplasmosis 2023)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>,<b>B</b>) The colonies on SDA at 25 °C showed white to buff-brown colored, suede-like to cottony mycelium on the recto and a pale brown reverse. (<b>C</b>) Microscopy analysis shows the presence of small, round to pyriform microconidia born on short branches on the sides of the hyphae at 20× (red arrows), scale bar 50 µm, and (<b>D</b>) characteristic large, rounded, single-celled, tuberculate macroconidia (black arrows) formed on short, hyaline, undifferentiated conidiophores at 100×, scale bar 10 µm.</p>
Full article ">
16 pages, 611 KiB  
Review
The State of the Art in Transcriptomics and Proteomics of Clinically Relevant Sporothrix Species
by Anna Carolina Procópio-Azevedo, Marcos de Abreu Almeida, Rodrigo Almeida-Paes, Rosely Maria Zancopé-Oliveira, Maria Clara Gutierrez-Galhardo, Priscila Marques de Macedo, Evandro Novaes, Alexandre Melo Bailão, Célia Maria de Almeida Soares and Dayvison Francis Saraiva Freitas
J. Fungi 2023, 9(8), 790; https://doi.org/10.3390/jof9080790 - 27 Jul 2023
Viewed by 1335
Abstract
Proteomics provide a robust approach to profile and quantify proteins within cells, organs, or tissues, providing comprehensive insights about the dynamics of cellular processes, modifications, and interactions. Similarly, understanding the transcriptome is essential to decipher functional elements of the genome, unraveling the mechanisms [...] Read more.
Proteomics provide a robust approach to profile and quantify proteins within cells, organs, or tissues, providing comprehensive insights about the dynamics of cellular processes, modifications, and interactions. Similarly, understanding the transcriptome is essential to decipher functional elements of the genome, unraveling the mechanisms of disease development and the molecular constituents of cells and tissues. Some thermodimorphic fungi of the genus Sporothrix cause sporotrichosis, a subcutaneous mycosis of worldwide relevance. The transcriptome and proteome of the main Sporothrix species of clinical interest can elucidate the mechanisms underlying pathogenesis and host interactions. Studies of these techniques can contribute to the advancement of novel diagnostic and therapeutic strategies. A literature review was carried out, addressing all articles based on proteomics using mass spectrometry and transcriptomics of Sporothrix spp. Twenty-one studies were eligible for this review. The main findings include proteins and genes involved in dimorphism, cell differentiation, thermotolerance, virulence, immune evasion, metabolism, cell adhesion, cell transport, and biosynthesis. With the spread and emergence of sporotrichosis in different countries, ongoing research efforts and new discoveries are welcome to advance knowledge about this mycosis and its agents. Full article
(This article belongs to the Section Fungal Pathogenesis and Disease Control)
Show Figures

Figure 1

Figure 1
<p>Flowchart detailing the steps of the articles search and those selected to be included in this study.</p>
Full article ">
10 pages, 2381 KiB  
Communication
Coccidioidal Pulmonary Cavitation: A New Age
by Lovedip Kooner, Augustine Munoz, Austin Garcia, Akriti Kaur, Rupam Sharma, Virginia Bustamante, Vishal Narang, George R. Thompson, Rasha Kuran, Amir Berjis, Royce H. Johnson and Arash Heidari
J. Fungi 2023, 9(5), 561; https://doi.org/10.3390/jof9050561 - 12 May 2023
Cited by 1 | Viewed by 1987
Abstract
Coccidioides species are thermally dimorphic fungi found in geographically defined areas of the Western Hemisphere. The primary portal of entry is respiratory, with symptomatic pneumonic diseases as the most common presentation. Subsequent pulmonary complications as well as extrapulmonary metastatic infection may occur, either [...] Read more.
Coccidioides species are thermally dimorphic fungi found in geographically defined areas of the Western Hemisphere. The primary portal of entry is respiratory, with symptomatic pneumonic diseases as the most common presentation. Subsequent pulmonary complications as well as extrapulmonary metastatic infection may occur, either of which may be the presenting disease manifestation. Cavitary lung disease may be found incidentally or when investigating symptoms such as cough or hemoptysis. This study aims to explore the spectrum of coccidioidal cavities and the evaluation and management in a cohort of patients seen at Kern Medical over the last 12 years. Full article
(This article belongs to the Special Issue Basic and Clinical Research on Coccidioides)
Show Figures

Figure 1

Figure 1
<p>Flowchart of patients with inclusion and exclusion criteria in the study of pulmonary cavitary coccidioidomycosis.</p>
Full article ">Figure 2
<p>Comparison of presenting manifestations for patients with coccidioidal pulmonary cavitations.</p>
Full article ">Figure 3
<p>Comparison of demographics for patients with coccidioidal pulmonary cavitations.</p>
Full article ">Figure 4
<p>CT image of a single coccidioidal pulmonary cavity. The red arrow indicates a cavity.</p>
Full article ">Figure 5
<p>X-ray image of a single coccidioidal pulmonary cavity. The red arrow indicates a cavity.</p>
Full article ">Figure 6
<p>CT image of multiple coccidioidal pulmonary cavities. The red arrows indicate cavities.</p>
Full article ">Figure 7
<p>X-ray image of multiple coccidioidal pulmonary cavities. The red arrows indicate cavities.</p>
Full article ">
14 pages, 854 KiB  
Review
Relationship of Sporotrichosis and Infected Patients with HIV-AIDS: An Actual Systematic Review
by Rodolfo Pinto-Almazán, Karla A. Sandoval-Navarro, Erika J. Damián-Magaña, Roberto Arenas, Claudia Erika Fuentes-Venado, Paola Berenice Zárate-Segura, Erick Martínez-Herrera and Carmen Rodríguez-Cerdeira
J. Fungi 2023, 9(4), 396; https://doi.org/10.3390/jof9040396 - 23 Mar 2023
Cited by 2 | Viewed by 2073
Abstract
Background: Sporotrichosis is a fungal infection that can affect both humans and animals, caused by a species of thermo-dimorphic fungi of the genus Sporothrix. This pathology can be acquired by subcutaneous traumatic inoculation through contact with contaminated plants, soil or decomposing organic [...] Read more.
Background: Sporotrichosis is a fungal infection that can affect both humans and animals, caused by a species of thermo-dimorphic fungi of the genus Sporothrix. This pathology can be acquired by subcutaneous traumatic inoculation through contact with contaminated plants, soil or decomposing organic matter, and/or by inhalation of conidia. The infection can progress to chronic skin infection, or it can even spread to blood vessels, lymph, muscles, bones, and other organs, such as the lungs and nervous system. Those disseminated types are usually associated with cellular immunodeficiency and infection by inhalation, which explains why people living with human immunodeficiency virus (PLHIV) get infected in such a manner. This virus changes the natural history of sporotrichosis, producing a greater fungal load. Methods: The search was carried out in three databases: Pubmed, Scopus, and Scielo. Eligible articles were considered as those that described sporotrichosis in patients infected with HIV-AIDS, as well as case series. Results: A total of 24 articles were selected, with a sum of 37 patients with sporotrichosis and HIV infection. Of these patients, 31 came from Brazil, two from the United States, one from South Africa, one from Bangladesh, and two from an unspecified region. Regarding epidemiology, a predominance of the male sex was found in 28 of the 37 cases (75.6%), while nine were female (24.3%). Conclusions: Sporotrichosis infection continues to present in a more severe and disseminated way among HIV-positive subjects with lower CD4+ counts. Full article
(This article belongs to the Special Issue Sporothrix and Sporotrichosis 3.0)
Show Figures

Figure 1

Figure 1
<p>Common characteristics of Sporotrichosis. (<b>A</b>) <span class="html-italic">Sporothrix</span> chancre, (<b>B</b>) <span class="html-italic">Sporothrix</span> yeasts on PAS-stained smears, (<b>C</b>) culture of <span class="html-italic">Sporothrix</span> spp. on Sabouraud dextrose agar with folded appearance.</p>
Full article ">Figure 2
<p>PRISMA flow diagram of data extracted from bibliographic searches.</p>
Full article ">
Back to TopTop