Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (201)

Search Parameters:
Keywords = malaria-infected cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
10 pages, 1975 KiB  
Communication
Measuring Asymmetric Ionic Current Waveform Through Micropores for Detecting Reduced Red Blood Cell Deformability Due to Plasmodium falciparum Infection
by Kazumichi Yokota, Ken Hirano, Kazuaki Kajimoto and Muneaki Hashimoto
Sensors 2025, 25(15), 4722; https://doi.org/10.3390/s25154722 - 31 Jul 2025
Viewed by 164
Abstract
The mechanisms underlying reduced deformability of red blood cells (RBCs) in Plasmodium falciparum remain unclear. The decrease in RBC deformability associated with malarial infection was measured using ektacytometry, and only mean values were evaluated. In this study, we report the development of a [...] Read more.
The mechanisms underlying reduced deformability of red blood cells (RBCs) in Plasmodium falciparum remain unclear. The decrease in RBC deformability associated with malarial infection was measured using ektacytometry, and only mean values were evaluated. In this study, we report the development of a microfluidic sensing device that can evaluate decreased RBC deformability at the single-cell level by measuring ionic current waveforms through micropores. Using an in vitro culture system, we found that when RBC deformability was reduced by P. falciparum infection, ionic current waveforms changed. As RBC deformability decreased, waveforms became asymmetric. Computer simulations suggested that these waveform parameters are largely independent of RBC size and may represent a reliable indicator of diminished deformability. This novel microfluidic RBC deformability sensor allows for detailed single-cell analysis of malaria-associated deformability reduction, potentially aiding in elucidating its pathology. Full article
(This article belongs to the Special Issue Recent Advances in Microfluidic Sensing Devices)
Show Figures

Graphical abstract

14 pages, 2113 KiB  
Article
NR2F6 as a Disease Driver and Candidate Therapeutic Target in Experimental Cerebral Malaria
by Victoria E. Stefan, Victoria Klepsch, Nikolaus Thuille, Martina Steinlechner, Sebastian Peer, Kerstin Siegmund, Peter Lackner, Erich Schmutzhard, Karin Albrecht-Schgör and Gottfried Baier
Cells 2025, 14(15), 1162; https://doi.org/10.3390/cells14151162 - 28 Jul 2025
Viewed by 256
Abstract
Cerebral malaria (CM) is the severe progression of an infection with Plasmodium falciparum, causing detrimental damage to brain tissue and is the most frequent cause of Plasmodium falciparum mortality. The critical role of brain-infiltrating CD8+ T cells in the pathophysiology of [...] Read more.
Cerebral malaria (CM) is the severe progression of an infection with Plasmodium falciparum, causing detrimental damage to brain tissue and is the most frequent cause of Plasmodium falciparum mortality. The critical role of brain-infiltrating CD8+ T cells in the pathophysiology of CM having been revealed, our investigation focuses on the role of NR2F6, an established immune checkpoint, as a candidate driver of CM pathology. We employed an experimental mouse model of CM based on Plasmodium berghei ANKA (PbA) infection to compare the relative susceptibility of Nr2f6-knock-out and wild-type C57BL6/N mice. As a remarkable result, Nr2f6 deficiency confers a significant survival benefit. In terms of mechanism, we detected less severe endotheliopathy and, hence, less damage to the blood–brain barrier (BBB), accompanied by decreased sequestered parasites and less cytotoxic T-lymphocytes within the brain, manifesting in a better disease outcome. We present evidence that NR2F6 deficiency renders mice more resistant to experimental cerebral malaria (ECM), confirming a causal and non-redundant role for NR2F6 in the progression of ECM disease. Consequently, pharmacological inhibitors of the NR2F6 pathway could be of use to bolster BBB integrity and protect against CM. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

18 pages, 2850 KiB  
Article
An mRNA Vaccine Expressing Blood-Stage Malaria Antigens Induces Complete Protection Against Lethal Plasmodium yoelii
by Amy C. Ott, Patrick J. Loll and James M. Burns
Vaccines 2025, 13(7), 702; https://doi.org/10.3390/vaccines13070702 - 28 Jun 2025
Viewed by 800
Abstract
Background and Objectives: To evaluate the mRNA vaccine platform for blood-stage Plasmodium parasites, we completed a proof-of-concept study using the P. yoelii mouse model of malaria and two mRNA-based vaccines. Both encoded PyMSP119 fused to PyMSP8 (PyMSP1/8). One [...] Read more.
Background and Objectives: To evaluate the mRNA vaccine platform for blood-stage Plasmodium parasites, we completed a proof-of-concept study using the P. yoelii mouse model of malaria and two mRNA-based vaccines. Both encoded PyMSP119 fused to PyMSP8 (PyMSP1/8). One was designed for secretion of the encoded protein (PyMSP1/8-sec); the other encoded membrane-bound antigen (PyMSP1/8-mem). Methods: Secretion of PyMSP1/8-sec and membrane localization of PyMSP1/8-mem were verified in mRNA-transfected cells. As recombinant PyMSP1/8 (rPyMSP1/8) is known to protect mice against lethal P. yoelii 17XL infection, we first compared immunogenicity and efficacy of the PyMSP1/8-sec mRNA vaccine versus the recombinant formulation in outbred mice. Animals were immunized three times followed by challenge with a lethal dose of P. yoelii 17XL-parasitized RBCs (pRBCs). Similar immunization and challenge experiments were conducted to compare PyMSP1/8-sec versus PyMSP1/8-mem mRNA vaccines. Results: Immunogenicity of the PyMSP1/8-sec mRNA vaccine was superior to the recombinant formulation, inducing higher antibody titers against both vaccine components. Following challenge with P. yoelii 17XL pRBCs, all PyMSP1/8-sec-immunized animals survived, with 50% of these showing no detectible pRBCs in circulation (<0.01%). In addition, mean peak parasitemia in PyMSP1/8-sec mRNA-immunized mice was significantly lower than that in the rPyMSP1/8 vaccine group. Both PyMSP1/8-sec and PyMSP1/8-mem were protective against P. yoelii 17XL challenge, with PyMSP1/8-mem immunization providing a significantly higher level of protection than PyMSP1/8-sec immunization considering the number of animals with no detectable pRBCs in circulation and the mean peak parasitemia in animals with detectable parasitemia. Conclusions: mRNA vaccines were highly immunogenic and potently protective against blood-stage malaria, outperforming a similar recombinant-based vaccine. The membrane-bound antigen was more effective at inducing protective antibody responses, highlighting the need to consider antigen localization for mRNA vaccine design. Full article
Show Figures

Figure 1

43 pages, 1769 KiB  
Review
The Role of LAIR1 as a Regulatory Receptor of Antitumor Immune Cell Responses and Tumor Cell Growth and Expansion
by Alessandro Poggi, Serena Matis, Chiara Rosa Maria Uras, Lizzia Raffaghello, Roberto Benelli and Maria Raffaella Zocchi
Biomolecules 2025, 15(6), 866; https://doi.org/10.3390/biom15060866 - 13 Jun 2025
Viewed by 842
Abstract
It is becoming evident that the therapeutic effect of reawakening the immune response is to limit tumor cell growth and expansion. The use of immune checkpoint inhibitors, like blocking antibodies against programmed cell death receptor (PD) 1 and/or cytotoxic T lymphocyte antigen (CTLA) [...] Read more.
It is becoming evident that the therapeutic effect of reawakening the immune response is to limit tumor cell growth and expansion. The use of immune checkpoint inhibitors, like blocking antibodies against programmed cell death receptor (PD) 1 and/or cytotoxic T lymphocyte antigen (CTLA) 4 alone or in combination with other drugs, has led to unexpected positive results in some tumors but not all. Several other molecules inhibiting lymphocyte antitumor effector subsets have been discovered in the last 30 years. Herein, we focus on the leukocyte-associated immunoglobulin (Ig)-like receptor 1 (LAIR1/CD305). LAIR1 represents a typical immunoregulatory molecule expressed on almost all leukocytes, unlike other regulatory receptors expressed on discrete leukocyte subsets. It bears two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in the intracytoplasmic protein domain involved in the downregulation of signals mediated by activating receptors. LAIR1 binds to several ligands, such as collagen I and III, complement component 1Q, surfactant protein D, adiponectin, and repetitive interspersed families of polypeptides expressed by erythrocytes infected with Plasmodium malariae. This would suggest LAIR1 involvement in several cell-to-cell interactions and possibly in metabolic regulation. The presence of both cellular and soluble forms of LAIR would indicate a fine regulation of the immunoregulatory activity, as happens for the soluble/exosome-associated forms of PD1 and CTLA4 molecules. As a consequence, LAIR1 appears to play a role in some autoimmune diseases and the immune response against tumor cells. The finding of LAIR1 expression on hematological malignancies, but also on some solid tumors, could open a rationale for the targeting of this molecule to treat neoplasia, either alone or in combination with other therapeutic options. Full article
Show Figures

Figure 1

4 pages, 1621 KiB  
Interesting Images
Encephalitis Unraveled: The Unlikely Encounter of Sickle Cell Disease and Cerebral Malaria in a Teenager
by Christer Ruff, Leonie Zerweck, Andrea Bevot, Jonathan Remppis, Benjamin Bender, Ulrike Ernemann and Georg Gohla
Diagnostics 2025, 15(12), 1470; https://doi.org/10.3390/diagnostics15121470 - 10 Jun 2025
Viewed by 427
Abstract
Sickle-cell disease (SCD) is a group of inherited blood disorders in which a mutation in the β-globin (HBB) gene causes red blood cells to produce abnormal hemoglobin, known as Hb S. SCD is characterized by an autosomal-recessive pattern of inheritance, implying that for [...] Read more.
Sickle-cell disease (SCD) is a group of inherited blood disorders in which a mutation in the β-globin (HBB) gene causes red blood cells to produce abnormal hemoglobin, known as Hb S. SCD is characterized by an autosomal-recessive pattern of inheritance, implying that for a child to manifest the condition, they must inherit an Hb S allele from both parents (HbSS) or one Hb S allele and another β-globin variant, such as Hb C or β-thalassemia (HbSC, HbS/β-thal). It has been observed that (heterozygote) carriers of one copy of the sickle-cell trait (HbAS) are typically healthy and can even gain partial protection from severe malaria. The term “severe and complicated malaria” is delineated based on specific clinical and laboratory characteristics in the presence of Plasmodium falciparum parasitemia. The prevalent forms of severe malaria among African children include cerebral malaria, respiratory distress, and severe malaria anemia. Cerebral malaria is a rare complication of malaria infection and is associated with a high mortality rate. Full article
(This article belongs to the Collection Interesting Images)
Show Figures

Figure 1

16 pages, 3636 KiB  
Article
Neuronal Damage in Murine Experimental Cerebral Malaria, Implications for Neuronal Repair and Sequelae
by Monique F. Stins, Irene Gramaglia, Joyce Velez, Carlos A. Pardo and Henri van der Heyde
Cells 2025, 14(11), 807; https://doi.org/10.3390/cells14110807 - 30 May 2025
Viewed by 597
Abstract
Cerebral malaria (CM) is a deadly complication of P. falciparum infection. Although adults with CM have a higher mortality rate, CM affects mostly children under the age of 5 years. Neurological symptoms and signs include impaired consciousness, coma, seizures, and increased intracranial hypertension. [...] Read more.
Cerebral malaria (CM) is a deadly complication of P. falciparum infection. Although adults with CM have a higher mortality rate, CM affects mostly children under the age of 5 years. Neurological symptoms and signs include impaired consciousness, coma, seizures, and increased intracranial hypertension. Upon survival of a CM episode, persistent neurologic deficits occur in a subset of surviving children. These sequelae include recurrent seizures, behavioral deficits, loss of developmental milestones, learning disabilities and attention deficit hyperactivity disorder, which can remain with the survivors. The underlying neuropathology of these post CM neurologic sequelae are unclear. Therefore, we probed the extensive neuronal damage that occurs in an experimental murine model of cerebral malaria (eCM), focusing on the hippocampus. In addition, we explored responses of neuro-progenitor cells (NPC’s) and potential repair mechanisms. We report here that Plasmodium infection causes extensive neuronal damage in the hippocampus, characterized by a loss of neuronal NeuN and double cortin (DCX) immunostaining in eCM mice. On day 6 of eCM we also observed increased neurofilament light chain staining, indicative of neuronal fragmentation, which was accompanied by an increase in neurofilament light chain in CSF but not seen in plasma. A concomitant increase in the influx of neuroprogenitor cells in eCM was observed, suggesting ongoing neuronal repair. Full article
Show Figures

Figure 1

18 pages, 2563 KiB  
Article
PLASMOpred: A Machine Learning-Based Web Application for Predicting Antimalarial Small Molecules Targeting the Apical Membrane Antigen 1–Rhoptry Neck Protein 2 Invasion Complex
by Eugene Lamptey, Jessica Oparebea, Gabriel Anyaele, Belinda Ofosu, George Hanson, Patrick O. Sakyi, Odame Agyapong, Dominic S. Y. Amuzu, Whelton A. Miller, Samuel K. Kwofie and Henrietta Esi Mensah-Brown
Pharmaceuticals 2025, 18(6), 776; https://doi.org/10.3390/ph18060776 - 23 May 2025
Viewed by 895
Abstract
Objective: Falciparum malaria is a major global health concern, affecting more than half of the world’s population and causing over half a million deaths annually. Red cell invasion is a crucial step in the parasite’s life cycle, where the parasite invade human erythrocytes [...] Read more.
Objective: Falciparum malaria is a major global health concern, affecting more than half of the world’s population and causing over half a million deaths annually. Red cell invasion is a crucial step in the parasite’s life cycle, where the parasite invade human erythrocytes to sustain infection and ensure survival. Two parasite proteins, Apical Membrane Antigen 1 (AMA-1) and Rhoptry Neck Protein 2 (RON2), are involved in tight junction formation, which is an essential step in parasite invasion of the red blood cell. Targeting the AMA-1 and RON2 interaction with inhibitors halts the formation of the tight junction, thereby preventing parasite invasion, which is detrimental to parasite survival. This study leverages machine learning (ML) to predict potential small molecule inhibitors of the AMA-1–RON2 interaction, providing putative antimalaria compounds for further chemotherapeutic exploration. Method: Data was retrieved from the PubChem database (AID 720542), comprising 364,447 inhibitors and non-inhibitors of the AMA-1–RON2 interaction. The data was processed by computing Morgan fingerprints and divided into training and testing with an 80:20 ratio, and the classes in the training data were balanced using the Synthetic Minority Oversampling Technique. Five ML models developed comprised Random Forest (RF), Gradient Boost Machines (GBMs), CatBoost (CB), AdaBoost (AB) and Support Vector Machine (SVM). The performances of the models were evaluated using accuracy, F1 score, and receiver operating characteristic—area under the curve (ROC-AUC) and validated using held-out data and a y-randomization test. An applicability domain analysis was carried out using the Tanimoto distance with a threshold set at 0.04 to ascertain the sample space where the models predict with confidence. Results: The GBMs model emerged as the best, achieving 89% accuracy and a ROC-AUC of 92%. CB and RF had accuracies of 88% and 87%, and ROC-AUC scores of 93% and 91%, respectively. Conclusions: Experimentally validated inhibitors of the AMA-1–RON2 interaction could serve as starting blocks for the next-generation antimalarial drugs. The models were deployed as a web-based application, known as PLASMOpred. Full article
(This article belongs to the Special Issue Artificial Intelligence-Assisted Drug Discovery)
Show Figures

Figure 1

16 pages, 2753 KiB  
Review
The Multifaceted Role of STK35/STK35L1 in Human Diseases: A Time for Critical Appraisal
by Arpana Yadav, Kritika Gaur, Phulwanti Kumari Sharma, Pragya Gehlot, Saloni Bage, Mahesh Saini, Daniela Brünnert and Pankaj Goyal
Kinases Phosphatases 2025, 3(2), 12; https://doi.org/10.3390/kinasesphosphatases3020012 - 23 May 2025
Viewed by 832
Abstract
Dysregulation of protein kinases is associated with developmental defects and various human diseases. The human kinome comprises 518 kinases, including several orphan kinases whose functions remain to be fully characterized. The NKF4 family, which includes STK35L1 and PDIK1L, is one such uncharacterized kinase [...] Read more.
Dysregulation of protein kinases is associated with developmental defects and various human diseases. The human kinome comprises 518 kinases, including several orphan kinases whose functions remain to be fully characterized. The NKF4 family, which includes STK35L1 and PDIK1L, is one such uncharacterized kinase family. STK35L1, also known as Clik1, was initially identified as a nuclear kinase associated with actin fibers. Subsequent studies have demonstrated that STK35L1 plays critical roles in cellular processes such as cell cycle regulation, migration, angiogenesis, the DNA damage response, and related processes such as spermatogenesis. STK35L1 has also been implicated in various developmental processes and its knockout mice exhibited defects in the testis, ovary, and eye. STK35L1 acts as a central regulator of the fundamental cellular functions, and its dysregulation leads to various diseases. Research has established that STK35L1 regulates tumor growth and proliferation in cancers such as osteosarcoma, colorectal cancer, and acute myeloid leukemia. Notably, it also affects chemosensitivity in colorectal cancer and metabolism in acute myeloid leukemia. Additionally, STK35L1 is crucial for the infection of hepatocytes by Plasmodium sporozoites during the liver stage of Malaria. This review discusses the current understanding of STK35L1, highlighting its role in various diseases. Full article
Show Figures

Figure 1

11 pages, 1688 KiB  
Article
Computer Viewing Model for Classification of Erythrocytes Infected with Plasmodium spp. Applied to Malaria Diagnosis Using Optical Microscope
by Eduardo Rojas, Irene Cartas-Espinel, Priscila Álvarez, Matías Moris, Manuel Salazar, Rodrigo Boguen, Pablo Letelier, Lucia San Martín, Valeria San Martín, Camilo Morales and Neftalí Guzmán
Medicina 2025, 61(5), 940; https://doi.org/10.3390/medicina61050940 - 21 May 2025
Viewed by 513
Abstract
Background and Objectives: Malaria is a disease that can result in a variety of complications. Diagnosis is carried out by an optical microscope and depends on operator experience. The use of artificial intelligence to identify morphological patterns in erythrocytes would improve our diagnostic [...] Read more.
Background and Objectives: Malaria is a disease that can result in a variety of complications. Diagnosis is carried out by an optical microscope and depends on operator experience. The use of artificial intelligence to identify morphological patterns in erythrocytes would improve our diagnostic capability. The object of this study was therefore to establish computer viewing models able to classify blood cells infected with Plasmodium spp. to support malaria diagnosis by optical microscope. Materials and Methods: A total of 27,558 images of human blood sample extensions were obtained from a public data bank for analysis; half were of parasite-infected red cells (n = 13,779), and the other half were of uninfected erythrocytes (n = 13,779). Six models (five machine learning algorithms and one pre-trained for a convolutional neural network) were assessed, and the performance of each was measured using metrics like accuracy (A), precision (P), recall, F1 score, and area under the curve (AUC). Results: The model with the best performance was VGG-19, with an AUC of 98%, accuracy of 93%, precision of 92%, recall of 94%, and F1 score of 93%. Conclusions: Based on the results, we propose a convolutional neural network model (VGG-19) for malaria diagnosis that can be applied in low-complexity laboratories thanks to its ease of implementation and high predictive performance. Full article
(This article belongs to the Section Hematology and Immunology)
Show Figures

Figure 1

20 pages, 986 KiB  
Review
Past, Present, and Future of Viral Vector Vaccine Platforms: A Comprehensive Review
by Justin Tang, Md Al Amin and Jian L. Campian
Vaccines 2025, 13(5), 524; https://doi.org/10.3390/vaccines13050524 - 15 May 2025
Viewed by 2625
Abstract
Over the past several decades, viral vector-based vaccines have emerged as some of the most versatile and potent platforms in modern vaccinology. Their capacity to deliver genetic material encoding target antigens directly into host cells enables strong cellular and humoral immune responses, often [...] Read more.
Over the past several decades, viral vector-based vaccines have emerged as some of the most versatile and potent platforms in modern vaccinology. Their capacity to deliver genetic material encoding target antigens directly into host cells enables strong cellular and humoral immune responses, often superior to what traditional inactivated or subunit vaccines can achieve. This has accelerated their application to a wide array of pathogens and disease targets, from well-established threats like HIV and malaria to emerging infections such as Ebola, Zika, and SARS-CoV-2. The COVID-19 pandemic further highlighted the agility of viral vector platforms, with several adenovirus-based vaccines quickly authorized and deployed on a global scale. Despite these advances, significant challenges remain. One major hurdle is pre-existing immunity against commonly used vector backbones, which can blunt vaccine immunogenicity. Rare but serious adverse events, including vector-associated inflammatory responses and conditions like vaccine-induced immune thrombotic thrombocytopenia (VITT), have raised important safety considerations. Additionally, scaling up manufacturing, ensuring consistency in large-scale production, meeting rigorous regulatory standards, and maintaining equitable global access to these vaccines present profound logistical and ethical dilemmas. In response to these challenges, the field is evolving rapidly. Sophisticated engineering strategies, such as integrase-defective lentiviral vectors, insect-specific flaviviruses, chimeric capsids to evade neutralizing antibodies, and plug-and-play self-amplifying RNA approaches, seek to bolster safety, enhance immunogenicity, circumvent pre-existing immunity, and streamline production. Lessons learned from the COVID-19 pandemic and prior outbreaks are guiding the development of platform-based approaches designed for rapid deployment during future public health emergencies. This review provides an exhaustive, in-depth examination of the historical evolution, immunobiological principles, current platforms, manufacturing complexities, regulatory frameworks, known safety issues, and future directions for viral vector-based vaccines. Full article
(This article belongs to the Special Issue Strategies of Viral Vectors for Vaccine Development)
Show Figures

Figure 1

21 pages, 1127 KiB  
Article
Efficient Compression of Red Blood Cell Image Dataset Using Joint Deep Learning-Based Pattern Classification and Data Compression
by Zerin Nusrat, Md Firoz Mahmud and W. David Pan
Electronics 2025, 14(8), 1556; https://doi.org/10.3390/electronics14081556 - 11 Apr 2025
Viewed by 506
Abstract
Millions of people across the globe are affected by the life-threatening disease of Malaria. To achieve the remote screening and diagnosis of the disease, the rapid transmission of large-size microscopic images is necessary, thereby demanding efficient data compression techniques. In this paper, we [...] Read more.
Millions of people across the globe are affected by the life-threatening disease of Malaria. To achieve the remote screening and diagnosis of the disease, the rapid transmission of large-size microscopic images is necessary, thereby demanding efficient data compression techniques. In this paper, we argued that well-classified images might lead to higher overall compression of the images in the datasets. To this end, we investigated the novel approach of joint pattern classification and compression of microscopic red blood cell images. Specifically, we used deep learning models, including a vision transformer and convolutional autoencoders, to classify red blood cell images into normal and Malaria-infected patterns, prior to applying compression on the images classified into different patterns separately. We evaluated the impacts of varying classification accuracy on overall image compression efficiency. The results highlight the importance of the accurate classification of images in improving overall compression performance. We demonstrated that the proposed deep learning-based joint classification/compression method offered superior performance compared with traditional lossy compression approaches such as JPEG and JPEG 2000. Our study provides useful insights into how deep learning-based pattern classification could benefit data compression, which would be advantageous in telemedicine, where large-image-size reduction and high decoded image quality are desired. Full article
(This article belongs to the Special Issue Deep Learning-Based Image Restoration and Object Identification)
Show Figures

Figure 1

22 pages, 2380 KiB  
Systematic Review
A Systematic Review and Meta-Analysis of MIP-1α and MIP-1β Chemokines in Malaria in Relation to Disease Severity
by Saruda Kuraeiad, Kwuntida Uthaisar Kotepui, Aongart Mahittikorn, Nsoh Godwin Anabire, Frederick Ramirez Masangkay, Polrat Wilairatana, Kinley Wangdi and Manas Kotepui
Medicina 2025, 61(4), 676; https://doi.org/10.3390/medicina61040676 - 6 Apr 2025
Viewed by 782
Abstract
Background and Objectives: Macrophage inflammatory protein-1α (MIP-1α) and MIP-1β act as signaling molecules that recruit immune cells to sites of infection and inflammation. This study aimed to synthesize evidence on blood levels of MIP-1α and MIP-1β in Plasmodium-infected individuals and to [...] Read more.
Background and Objectives: Macrophage inflammatory protein-1α (MIP-1α) and MIP-1β act as signaling molecules that recruit immune cells to sites of infection and inflammation. This study aimed to synthesize evidence on blood levels of MIP-1α and MIP-1β in Plasmodium-infected individuals and to determine whether these levels differ between severe and uncomplicated malaria cases. Materials and Methods: The study protocol was registered in PROSPERO (CRD42024595818). Comprehensive literature searches were conducted in six databases (EMBASE, MEDLINE, Ovid, Scopus, ProQuest, and PubMed) to identify studies reporting blood levels of MIP-1α and MIP-1β in Plasmodium infections and clinical malaria. A narrative synthesis was used to describe variations in MIP-1α and MIP-1β levels between malaria patients and controls and between severe and non-severe malaria cases. Meta-analysis was used to aggregate quantitative data utilizing a random-effects model. Results: A total of 1638 records were identified, with 20 studies meeting the inclusion criteria. Most studies reported significantly higher MIP-1α and MIP-1β levels in malaria patients compared to non-malarial controls. The meta-analysis showed a significant elevation in MIP-1α levels in malaria patients (n = 352) compared to uninfected individuals (n = 274) (p = 0.0112, random effects model, standardized mean difference [SMD]: 1.69, 95% confidence interval [CI]: 0.38 to 3.00, I2: 96.0%, five studies, 626 individuals). The meta-analysis showed no difference in MIP-1α levels between severe malaria cases (n = 203) and uncomplicated cases (n = 106) (p = 0.51, SMD: −0.48, 95% CI: −1.93 to 0.96, I2: 97.3%, three studies, 309 individuals). Conclusions: This study suggests that while MIP-1α and MIP-1β levels are elevated in malaria patients compared to uninfected individuals, these chemokines show a limited ability to differentiate between severe and uncomplicated malaria or predict severe outcomes. Further research is needed to clarify their role in malaria pathogenesis and explore potential clinical applications. Full article
(This article belongs to the Special Issue Infectious and Tropical Diseases: Symptoms, Diagnosis and Treatment)
Show Figures

Figure 1

23 pages, 2184 KiB  
Article
Lossless Compression of Malaria-Infected Erythrocyte Images Using Vision Transformer and Deep Autoencoders
by Md Firoz Mahmud, Zerin Nusrat and W. David Pan
Computers 2025, 14(4), 127; https://doi.org/10.3390/computers14040127 - 1 Apr 2025
Viewed by 680
Abstract
Lossless compression of medical images allows for rapid image data exchange and faithful recovery of the compressed data for medical image assessment. There are many useful telemedicine applications, for example in diagnosing conditions such as malaria in resource-limited regions. This paper presents a [...] Read more.
Lossless compression of medical images allows for rapid image data exchange and faithful recovery of the compressed data for medical image assessment. There are many useful telemedicine applications, for example in diagnosing conditions such as malaria in resource-limited regions. This paper presents a novel machine learning-based approach where lossless compression of malaria-infected erythrocyte images is assisted by cutting-edge classifiers. To this end, we first use a Vision Transformer to classify images into two categories: those cells that are infected with malaria and those that are not. We then employ distinct deep autoencoders for each category, which not only reduces the dimensions of the image data but also preserves crucial diagnostic information. To ensure no loss in reconstructed image quality, we further compress the residuals produced by these autoencoders using the Huffman code. Simulation results show that the proposed method achieves lower overall bit rates and thus higher compression ratios than traditional compression schemes such as JPEG 2000, JPEG-LS, and CALIC. This strategy holds significant potential for effective telemedicine applications and can improve diagnostic capabilities in regions impacted by malaria. Full article
Show Figures

Graphical abstract

22 pages, 4045 KiB  
Article
Differential Response and Recovery Dynamics of HSPC Populations Following Plasmodium chabaudi Infection
by Federica Bruno, Christiana Georgiou, Deirdre Cunningham, Lucy Bett, Marine A. Secchi, Samantha Atkinson, Sara González Antón, Flora Birch, Jean Langhorne and Cristina Lo Celso
Int. J. Mol. Sci. 2025, 26(6), 2816; https://doi.org/10.3390/ijms26062816 - 20 Mar 2025
Viewed by 712
Abstract
Severe infections such as malaria are on the rise worldwide, driven by both climate change and increasing drug resistance. It is therefore paramount that we better understand how the host responds to severe infection. Hematopoiesis is particularly of interest in this context because [...] Read more.
Severe infections such as malaria are on the rise worldwide, driven by both climate change and increasing drug resistance. It is therefore paramount that we better understand how the host responds to severe infection. Hematopoiesis is particularly of interest in this context because hematopoietic stem and progenitor cells (HSPCs) maintain the turnover of all blood cells, including all immune cells. Severe infections have been widely acknowledged to affect HSPCs; however, this disruption has been mainly studied during the acute phase, and the process and level of HSPC recovery remain understudied. Using a self-resolving model of natural rodent malaria, infection by Plasmodium chabaudi, here we systematically assess phenotypically defined HSPCs’ acute response and recovery upon pathogen clearance. We demonstrate that during the acute phase of infection the most quiescent and functional stem cells are depleted, multipotent progenitor compartments are drastically enlarged, and oligopotent progenitors virtually disappear, underpinned by dramatic, population-specific and sometimes unexpected changes in proliferation rates. HSPC populations return to homeostatic size and proliferation rate again through specific patterns of recovery. Overall, our data demonstrate that HSPC populations adopt different responses to cope with severe infection and suggest that the ability to adjust proliferative capacity becomes more restricted as differentiation progresses. Full article
Show Figures

Graphical abstract

21 pages, 315 KiB  
Review
Unraveling the Role of Proteinopathies in Parasitic Infections
by Mikołaj Hurła, Damian Pikor, Natalia Banaszek-Hurła, Alicja Drelichowska, Jolanta Dorszewska, Wojciech Kozubski, Elżbieta Kacprzak and Małgorzata Paul
Biomedicines 2025, 13(3), 610; https://doi.org/10.3390/biomedicines13030610 - 3 Mar 2025
Viewed by 1287
Abstract
Proteinopathies, characterized by the misfolding, aggregation, and deposition of proteins, are hallmarks of various neurodegenerative and systemic diseases. Increasingly, research has highlighted the role of protein misfolding in parasitic infections, unveiling intricate interactions between host and parasite that exacerbate disease pathology and contribute [...] Read more.
Proteinopathies, characterized by the misfolding, aggregation, and deposition of proteins, are hallmarks of various neurodegenerative and systemic diseases. Increasingly, research has highlighted the role of protein misfolding in parasitic infections, unveiling intricate interactions between host and parasite that exacerbate disease pathology and contribute to chronic outcomes. The life cycles of parasitic protozoa, including Plasmodium, Toxoplasmosis, and Leishmania species, are complicated and involve frequent changes between host and vector environments. Their proteomes are severely stressed during these transitions, which calls for highly specialized protein quality control systems. In order to survive harsh intracellular conditions during infection, these parasites have been demonstrated to display unique adaptations in the unfolded protein response, a crucial pathway controlling endoplasmic reticulum stress. In addition to improving parasite survival, these adaptations affect host cell signaling and metabolism, which may jeopardize cellular homeostasis. By causing oxidative stress, persistent inflammation, and disturbance of cellular proteostasis, host–parasite interactions also contribute to proteinopathy. For instance, Plasmodium falciparum disrupts normal protein homeostasis and encourages the accumulation of misfolded proteins by influencing host redox systems involved in protein folding. In addition to interfering with host chaperone systems, the parasitic secretion of effector proteins exacerbates protein misfolding and aggregate formation. Autophagy, apoptosis regulation, organelle integrity, and other vital cellular processes are all disrupted by these pathological protein aggregates. Long-term misfolding and aggregation can cause irreversible tissue damage, which can worsen the clinical course of illnesses like visceral leishmaniasis, cerebral malaria, and toxoplasmosis. Treating parasite-induced proteinopathies is a potentially fruitful area of therapy. According to recent research, autophagy modulators, proteasome enhancers, and small-molecule chaperones may be repurposed to lessen these effects. Pharmacological agents that target the UPR, for example, have demonstrated the ability to decrease parasite survival while also reestablishing host protein homeostasis. Targeting the proteins secreted by parasites that disrupt host proteostasis may also offer a novel way to stop tissue damage caused by proteinopathies. In conclusion, the intersection of protein misfolding and parasitic infections represents a rapidly advancing field of research. Dissecting the molecular pathways underpinning these processes offers unprecedented opportunities for developing innovative therapies. These insights could not only transform the management of parasitic diseases but also contribute to a broader understanding of proteinopathies in infectious and non-infectious diseases alike. Full article
(This article belongs to the Special Issue Advanced Research in Proteinopathies)
Back to TopTop