1
|
Amer MM, Soliman AM, DO T, Hegab AA, El-Kelesh EA, Li Y, Jaroszewski J, Mohanta UK, Xuan X. Parasitological and molecular investigation of Trypanosoma evansi in dromedaries from Greater Cairo, Egypt. J Vet Med Sci 2024; 86:1177-1184. [PMID: 39358244 DOI: 10.1292/jvms.24-0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
In Egypt, camel trypanosomiasis is widespread. From October 2021 to March 2022, we collected 181 blood samples from apparently healthy one-humped camels (Camelus dromedarius) in Cairo and Giza Governates. The objective of this study was to assess infection rates of trypanosomes using blood smear examination and PCR-sequencing assays. Trypanosomes were detected in 8.3% (15/181) of camels by blood smear and in 23.8% (43/181) by PCR targeting the internal transcribed spacer (ITS). Based on blood smear and ITS-PCR results, and the absence of tsetse flies in the study area, we hypothesized that the Trypanosoma species was likely T. evansi. Validation using PCR based on the variant surface glycoprotein (VSG) of T. evansi Rode Trypanozoon antigen type (RoTat) 1.2 (RoTat 1.2 VSG gene) on ITS-PCR-positive samples (n=43) confirmed that 88.4% (38/43) were RoTat 1.2 T. evansi, while 11.6% (5/43) were non-RoTat 1.2 T. evansi. This marks the second report of non-RoTat 1.2 T. evansi in dromedary camels in Egypt. Considering the underestimated zoonotic risk of T. evansi in Egypt, there is a potential threat to humans, underscoring the need for a "One Health" approach to safeguard animal and human health.
Collapse
Affiliation(s)
- Moaz M Amer
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Department of Biotechnology, Animal Health Research Institute, Dokki, Egypt
| | - Ahmed M Soliman
- Department of Biotechnology, Animal Health Research Institute, Dokki, Egypt
| | - Thom DO
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | | | | | - Yongchang Li
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Jerzy Jaroszewski
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Uday Kumar Mohanta
- Department of Microbiology and Parasitology, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| |
Collapse
|
2
|
Pays E. Apolipoprotein-L1 (APOL1): From Sleeping Sickness to Kidney Disease. Cells 2024; 13:1738. [PMID: 39451256 PMCID: PMC11506758 DOI: 10.3390/cells13201738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Apolipoprotein-L1 (APOL1) is a membrane-interacting protein induced by inflammation, which confers human resistance to infection by African trypanosomes. APOL1 kills Trypanosoma brucei through induction of apoptotic-like parasite death, but two T. brucei clones acquired resistance to APOL1, allowing them to cause sleeping sickness. An APOL1 C-terminal sequence alteration, such as occurs in natural West African variants G1 and G2, restored human resistance to these clones. However, APOL1 unfolding induced by G1 or G2 mutations enhances protein hydrophobicity, resulting in kidney podocyte dysfunctions affecting renal filtration. The mechanism involved in these dysfunctions is debated. The ability of APOL1 to generate ion pores in trypanosome intracellular membranes or in synthetic membranes was provided as an explanation. However, transmembrane insertion of APOL1 strictly depends on acidic conditions, and podocyte cytopathology mainly results from secreted APOL1 activity on the plasma membrane, which occurs under non-acidic conditions. In this review, I argue that besides inactivation of APOL3 functions in membrane dynamics (fission and fusion), APOL1 variants induce inflammation-linked podocyte toxicity not through pore formation, but through plasma membrane disturbance resulting from increased interaction with cholesterol, which enhances cation channels activity. A natural mutation in the membrane-interacting domain (N264K) abrogates variant APOL1 toxicity at the expense of slightly increased sensitivity to trypanosomes, further illustrating the continuous mutual adaptation between host and parasite.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| |
Collapse
|
3
|
Vaisar T, Babenko I, Horvath KV, Niisuke K, Asztalos BF. Relationships between HDL subpopulation proteome and HDL function in overweight/obese people with and without coronary heart disease. Atherosclerosis 2024; 397:118565. [PMID: 39260003 PMCID: PMC11539851 DOI: 10.1016/j.atherosclerosis.2024.118565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS The structure-function relationships of high-density lipoprotein (HDL) subpopulations are not well understood. Our aim was to examine the interrelationships between HDL particle proteome and HDL functionality in subjects with and without coronary heart disease (CHD). METHODS We isolated 5 different HDL subpopulations based on charge, size, and apolipoprotein A1 (APOA1) content from the plasma of 33 overweight/obese CHD patients and 33 age-and body mass index (BMI)-matched CHD-free subjects. We measured the relative molar concentration of HDL-associated proteins by liquid chromatography tandem mass spectrometry (LC-MS/MS) and assessed particle functionality. RESULTS We quantified 110 proteins associated with the 5 APOA1-containing HDL subpopulations. The relative molar concentration of these proteins spanned five orders of magnitude. Only 10 proteins were present in >1% while 73 were present in <0.1% concentration. Only 6 of the 10 most abundant proteins were apolipoproteins. Interestingly, the largest (α-1) and the smallest (preβ-1) HDL particles contained the most diverse proteomes. The protein composition of each HDL subpopulation was altered in CHD cases as compared to controls with the most prominent differences in preβ-1 and α-1 particles. APOA2 concentration was positively correlated with preβ-1 particle functionality (ABCA1-CEC/mg APOA1 in preβ-1) (R2 = 0.42, p = 0.005), while APOE concentration was inversely correlated with large-HDL particle functionality (SRBI-CEC/mg APOA1 in α-1+α-2) (R2 = 0.18, p = 0.01). CONCLUSIONS The protein composition of the different HDL subpopulations was altered differentially in CHD patients. The functionality of the small and large HDL particles correlated with the protein content of APOA2 and APOE, respectively. Our data indicate that distinct particle subspecies and specific particle associated proteins provide new information about the role of HDL in CHD.
Collapse
Affiliation(s)
- Tomas Vaisar
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Ilona Babenko
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Katalin V Horvath
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Katrin Niisuke
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Bela F Asztalos
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
4
|
Gulati A, Moxey-Mims M. Defining Risk in APOL1-Associated Kidney Disease: The Story is Evolving! Am J Kidney Dis 2024; 84:388-391. [PMID: 38648881 DOI: 10.1053/j.ajkd.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Ashima Gulati
- Division of Pediatric Nephrology, Children's National Hospital, Washington, DC; Department of Pediatrics, The George Washington University School of Medicine, Washington, DC
| | - Marva Moxey-Mims
- Division of Pediatric Nephrology, Children's National Hospital, Washington, DC; Department of Pediatrics, The George Washington University School of Medicine, Washington, DC.
| |
Collapse
|
5
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
6
|
Hubacek JA, Adamkova V, Lanska V, Stanek V, Mrazkova J, Gebauerova M, Kettner J, Kautzner J, Pitha J. APOL1 polymorphisms are not influencing acute coronary syndrome risk in Czech males. Mol Genet Genomic Med 2024; 12:e2449. [PMID: 39171649 PMCID: PMC11339648 DOI: 10.1002/mgg3.2449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND The highest mortality and morbidity worldwide is associated with atherosclerotic cardiovascular disease (ASCVD), which has in background both environmental and genetic risk factors. Apolipoprotein L1 (APOL1) variability influences the risk of ASCVD in Africans, but little is known about the APOL1 and ASCVD in other ethnic groups. METHODS To investigate the role of APOL1 and ASCVD, we have genotyped four (rs13056427, rs136147, rs10854688 and rs9610473) APOL1 polymorphisms in a group of 1541 male patients with acute coronary syndrome (ACS) and 1338 male controls. RESULTS Individual APOL1 polymorphisms were not associated with traditional CVD risk factors such as smoking, hypertension or diabetes prevalence, with BMI values or plasma lipid levels. Neither individual polymorphisms nor haplotypes were associated with an increased risk of ACS nor did they predict total or cardiovascular mortality over the 10.2 ± 3.9 years of follow-up. CONCLUSIONS We conclude that APOL1 genetic variability has no major effect on risk of ACS in Caucasians.
Collapse
Affiliation(s)
- Jaroslav A. Hubacek
- Experimental Medicine CentreInstitute for Clinical and Experimental MedicinePragueCzech Republic
- 3rd Department of Internal Medicine, 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Vera Adamkova
- Preventive Cardiology CentreInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Vera Lanska
- Department of InformaticsInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Vladimir Stanek
- Cardiology DepartmentInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Jolana Mrazkova
- Experimental Medicine CentreInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Marie Gebauerova
- Cardiology DepartmentInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Jiri Kettner
- Cardiology DepartmentInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Josef Kautzner
- Cardiology DepartmentInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Jan Pitha
- Experimental Medicine CentreInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
7
|
Banerjee S, Minshall N, Webb H, Carrington M. How are Trypanosoma brucei receptors protected from host antibody-mediated attack? Bioessays 2024; 46:e2400053. [PMID: 38713161 DOI: 10.1002/bies.202400053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024]
Abstract
Trypanosoma brucei is the causal agent of African Trypanosomiasis in humans and other animals. It maintains a long-term infection through an antigenic variation based population survival strategy. To proliferate in a mammal, T. brucei acquires iron and haem through the receptor mediated uptake of host transferrin and haptoglobin-hemoglobin respectively. The receptors are exposed to host antibodies but this does not lead to clearance of the infection. Here we discuss how the trypanosome avoids this fate in the context of recent findings on the structure and cell biology of the receptors.
Collapse
Affiliation(s)
- Sourav Banerjee
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Nicola Minshall
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Helena Webb
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Mark Carrington
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Olono A, Mitesser V, Happi A, Happi C. Building genomic capacity for precision health in Africa. Nat Med 2024; 30:1856-1864. [PMID: 38961224 DOI: 10.1038/s41591-024-03081-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024]
Abstract
The African continent is poised to have a pivotal role in the global population landscape, with the United Nations projecting a population of 2.5 billion (more than 25% of the global population) by 2050. Amid this demographic shift, Africa faces a unique healthcare challenge-navigating a complex landscape of infectious and non-communicable diseases. This necessitates a departure from the conventional 'one-size-fits-all' medical model toward precision approaches that are efficient and sustainable. Genomic capacity is a pillar of precision health; however, access to up-to-date genetic testing in African countries is limited, compounded by a startling lack of representation of data from populations of African descent in gene discovery studies. In this Review, we delve into the challenges impeding the development of genomic capacity in Africa, such as the lack of electronic clinical and epidemiological records, infrastructural challenges, high supply chain costs and the 'dependency trap' that jeopardizes long-term sustainability. We emphasize the need for strategies hinged on true partnerships, robust infrastructure, workforce development and well-crafted policies. Finally, we outline recent progress and existing initiatives that should be considered as role models for future capacity-building initiatives.
Collapse
Affiliation(s)
- Alhaji Olono
- The African Centre of Excellence for Genomics and Infectious Diseases, ACEGID, Ede, Nigeria
| | - Vera Mitesser
- The African Centre of Excellence for Genomics and Infectious Diseases, ACEGID, Ede, Nigeria
| | - Anise Happi
- The African Centre of Excellence for Genomics and Infectious Diseases, ACEGID, Ede, Nigeria
| | - Christian Happi
- The African Centre of Excellence for Genomics and Infectious Diseases, ACEGID, Ede, Nigeria.
| |
Collapse
|
9
|
Dogan M, Watkins C, Ingram H, Moore N, Rucker GM, Gower EG, Eason JD, Bhalla A, Talwar M, Nezakatgoo N, Eymard C, Helmick R, Vanatta J, Bajwa A, Kuscu C, Kuscu C. Unveiling APOL1 haplotypes in a predominantly African-American cohort of kidney transplant patients: a novel classification using probe-independent quantitative real-time PCR. Front Med (Lausanne) 2024; 11:1325128. [PMID: 38660426 PMCID: PMC11039853 DOI: 10.3389/fmed.2024.1325128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Apolipoprotein-L1 (APOL1) is a primate-specific protein component of high-density lipoprotein (HDL). Two variants of APOL1 (G1 and G2), provide resistance to parasitic infections in African Americans but are also implicated in kidney-related diseases and transplant outcomes in recipients. This study aims to identify these risk variants using a novel probe-independent quantitative real-time PCR method in a high African American recipient cohort. Additionally, it aims to develop a new stratification approach based on a haplotype-centric model. Methods Genomic DNA was extracted from recipient PBMCs using SDS lysis buffer and proteinase K. A quantitative PCR assay with modified forward primers and a common reverse primer enabled us to quantitatively identify single nucleotide polymorphisms (SNPs) and the 6-bp deletion. Additionally, we used Sanger sequencing to verify our QPCR findings. Results Our novel probe-independent qPCR effectively distinguished homozygous wild-type, heterozygous SNPs/deletions, and homozygous SNPs/deletions, with at least 4-fold differences. A high prevalence of APOL1 variants was observed (18% two-risk alleles, 34% one-risk allele) in our recipient cohort. Intriguingly, no significant impact of recipient APOL1 variants on transplant outcomes was observed up to 12-month of follow-ups. Ongoing research will encompass more time points and a larger patient cohort, allowing for a comprehensive evaluation of G1/G2 variant subgroups categorized by new haplotype scores, enriching our understanding. Conclusion Our cost-effective and rapid qPCR technique facilitates APOL1 genotyping within hours. Prospective and retrospective studies will enable comparisons with long-term allograft rejection, potentially predicting early/late-stage transplant outcomes based on haplotype evaluation in this diverse group of kidney transplant recipients.
Collapse
Affiliation(s)
- Murat Dogan
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Christine Watkins
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Holly Ingram
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nicholas Moore
- Transplant Research Institute, Memphis, TN, United States
| | - Grace M. Rucker
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | | | | | - Anshul Bhalla
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Manish Talwar
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Nosratollah Nezakatgoo
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Corey Eymard
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Ryan Helmick
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Jason Vanatta
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Amandeep Bajwa
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Canan Kuscu
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Cem Kuscu
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
10
|
Pays E. The Janus-faced functions of Apolipoproteins L in membrane dynamics. Cell Mol Life Sci 2024; 81:134. [PMID: 38478101 PMCID: PMC10937811 DOI: 10.1007/s00018-024-05180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/18/2024] [Indexed: 03/17/2024]
Abstract
The functions of human Apolipoproteins L (APOLs) are poorly understood, but involve diverse activities like lysis of bloodstream trypanosomes and intracellular bacteria, modulation of viral infection and induction of apoptosis, autophagy, and chronic kidney disease. Based on recent work, I propose that the basic function of APOLs is the control of membrane dynamics, at least in the Golgi and mitochondrion. Together with neuronal calcium sensor-1 (NCS1) and calneuron-1 (CALN1), APOL3 controls the activity of phosphatidylinositol-4-kinase-IIIB (PI4KB), involved in both Golgi and mitochondrion membrane fission. Whereas secreted APOL1 induces African trypanosome lysis through membrane permeabilization of the parasite mitochondrion, intracellular APOL1 conditions non-muscular myosin-2A (NM2A)-mediated transfer of PI4KB and APOL3 from the Golgi to the mitochondrion under conditions interfering with PI4KB-APOL3 interaction, such as APOL1 C-terminal variant expression or virus-induced inflammatory signalling. APOL3 controls mitophagy through complementary interactions with the membrane fission factor PI4KB and the membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). In mice, the basic APOL1 and APOL3 activities could be exerted by mAPOL9 and mAPOL8, respectively. Perspectives regarding the mechanism and treatment of APOL1-related kidney disease are discussed, as well as speculations on additional APOLs functions, such as APOL6 involvement in adipocyte membrane dynamics through interaction with myosin-10 (MYH10).
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041, Gosselies, Belgium.
| |
Collapse
|
11
|
Gupta N, Waas B, Austin D, De Mazière AM, Kujala P, Stockwell AD, Li T, Yaspan BL, Klumperman J, Scales SJ. Apolipoprotein L1 (APOL1) renal risk variant-mediated podocyte cytotoxicity depends on African haplotype and surface expression. Sci Rep 2024; 14:3765. [PMID: 38355600 PMCID: PMC10866943 DOI: 10.1038/s41598-024-53298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Homozygous Apolipoprotein L1 (APOL1) variants G1 and G2 cause APOL1-mediated kidney disease, purportedly acting as surface cation channels in podocytes. APOL1-G0 exhibits various single nucleotide polymorphisms, most commonly haplotype E150K, M228I and R255K ("KIK"; the Reference Sequence is "EMR"), whereas variants G1 and G2 are mostly found in a single "African" haplotype background ("EIK"). Several labs reported cytotoxicity with risk variants G1 and G2 in KIK or EIK background haplotypes, but used HEK-293 cells and did not verify equal surface expression. To see if haplotype matters in a more relevant cell type, we induced APOL1-G0, G1 and G2 EIK, KIK and EMR at comparable surface levels in immortalized podocytes. G1 and G2 risk variants (but not G0) caused dose-dependent podocyte death within 48h only in their native African EIK haplotype and correlated with K+ conductance (thallium FLIPR). We ruled out differences in localization and trafficking, except for possibly greater surface clustering of cytotoxic haplotypes. APOL1 surface expression was required, since Brefeldin A rescued cytotoxicity; and cytoplasmic isoforms vB3 and vC were not cytotoxic. Thus, APOL1-EIK risk variants kill podocytes in a dose and haplotype-dependent manner (as in HEK-293 cells), whereas unlike in HEK-293 cells the KIK risk variants did not.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Discovery Immunology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Bridget Waas
- Department of Discovery Immunology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel Austin
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Ann M De Mazière
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pekka Kujala
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Amy D Stockwell
- Department of Human Genetics, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Tianbo Li
- Department of Biochemical and Cellular Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Brian L Yaspan
- Department of Human Genetics, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Judith Klumperman
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Suzie J Scales
- Department of Discovery Immunology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
12
|
Hu Q, Tong Z, Yalikong A, Ge LP, Shi Q, Du X, Wang P, Liu XY, Zhan W, Gao X, Sun D, Fu T, Ye D, Fan C, Liu J, Zhong YS, Jiang YZ, Gu H. DNAzyme-based faithful probing and pulldown to identify candidate biomarkers of low abundance. Nat Chem 2024; 16:122-131. [PMID: 37710046 DOI: 10.1038/s41557-023-01328-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/17/2023] [Indexed: 09/16/2023]
Abstract
Biomarker discovery is essential for the understanding, diagnosis, targeted therapy and prognosis assessment of malignant diseases. However, it remains a huge challenge due to the lack of sensitive methods to identify disease-specific rare molecules. Here we present MORAC, molecular recognition based on affinity and catalysis, which enables the effective identification of candidate biomarkers with low abundance. MORAC relies on a class of DNAzymes, each cleaving a sole RNA linkage embedded in their DNA chain upon specifically sensing a complex system with no prior knowledge of the system's molecular content. We show that signal amplification from catalysis ensures the DNAzymes high sensitivity (for target probing); meanwhile, a simple RNA-to-DNA mutation can shut down their RNA cleavage ability and turn them into a pure affinity tool (for target pulldown). Using MORAC, we identify previously unknown, low-abundance candidate biomarkers with clear clinical value, including apolipoprotein L6 in breast cancer and seryl-tRNA synthetase 1 in polyps preceding colon cancer.
Collapse
Affiliation(s)
- Qinqin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, and School of Global Health, Shanghai Jiao Tong University, Shanghai, China
| | - Zongxuan Tong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ayimukedisi Yalikong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li-Ping Ge
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Shi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyu Du
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pu Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi-Yu Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wuqiang Zhan
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xia Gao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Di Sun
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tong Fu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan Ye
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunhai Fan
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, and School of Global Health, Shanghai Jiao Tong University, Shanghai, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Laboratory, Shanghai, China
| | - Jie Liu
- Department of Digestive Disease, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun-Shi Zhong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hongzhou Gu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Institutes of Biomedical Sciences, Fudan University Shanghai Cancer Center, Shanghai Stomatological Hospital, and Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, and School of Global Health, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Janse van Rensburg HD, N'Da DD, Suganuma K. In vitro trypanocidal potency and in vivo treatment efficacy of oligomeric ethylene glycol-tethered nitrofurantoin derivatives. Eur J Pharm Sci 2024; 192:106668. [PMID: 38065268 DOI: 10.1016/j.ejps.2023.106668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023]
Abstract
African trypanosomiasis is a significant vector-borne disease of humans and animals in the tsetse fly belt of Africa, particularly affecting production animals such as cattle, and thus, hindering food security. Trypanosoma congolense (T. congolense), the causative agent of nagana, is livestock's most virulent trypanosome species. There is currently no vaccine against trypanosomiasis; its treatment relies solely on chemotherapy. However, pathogenic resistance has been established against trypanocidal agents in clinical use. This underscores the need to develop new therapeutics to curb trypanosomiasis. Many nitroheterocyclic drugs or compounds, including nitrofurantoin, possess antiparasitic activities in addition to their clinical use as antibiotics. The current study evaluated the in vitro trypanocidal potency and in vivo treatment efficacy of previously synthesized antileishmanial active oligomeric ethylene glycol derivatives of nitrofurantoin. The trypanocidal potency of analogues 2a-o varied among the trypanosome species; however, T. congolense strain IL3000 was more susceptible to these drug candidates than the other human and animal trypanosomes. The arylated analogues 2k (IC50 0.04 µM; SI >6365) and 2l (IC50 0.06 µM; SI 4133) featuring 4-chlorophenoxy and 4-nitrophenoxy moieties, respectively, were revealed as the most promising antitrypanosomal agents of all analogues against T. congolense strain IL3000 trypomastigotes with nanomolar activities. In a preliminary in vivo study involving T. congolense strain IL3000 infected BALB/c mice, the oral administration of 100 mg/kg/day of 2k caused prolonged survival up to 18 days post-infection relative to the infected but untreated control mice which survived 9 days post-infection. However, no cure was achieved due to its poor solubility in the in vivo testing medium, assumably leading to low oral bioavailability. These results confirm the importance of the physicochemical properties lipophilicity and water solubility in attaining not only in vitro trypanocidal potency but also in vivo treatment efficacy. Future work will focus on the chemical optimization of 2k through the investigation of analogues containing solubilizing groups at certain positions on the core structure to improve solubility in the in vivo testing medium which, in the current investigation, is the biggest stumbling block in successfully treating either animal or human Trypanosoma infections.
Collapse
Affiliation(s)
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
14
|
Lecordier L, Heo P, Graversen JH, Hennig D, Skytthe MK, Cornet d'Elzius A, Pincet F, Pérez-Morga D, Pays E. Apolipoproteins L1 and L3 control mitochondrial membrane dynamics. Cell Rep 2023; 42:113528. [PMID: 38041817 PMCID: PMC10765320 DOI: 10.1016/j.celrep.2023.113528] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023] Open
Abstract
Apolipoproteins L1 and L3 (APOLs) are associated at the Golgi with the membrane fission factors phosphatidylinositol 4-kinase-IIIB (PI4KB) and non-muscular myosin 2A. Either APOL1 C-terminal truncation (APOL1Δ) or APOL3 deletion (APOL3-KO [knockout]) reduces PI4KB activity and triggers actomyosin reorganization. We report that APOL3, but not APOL1, controls PI4KB activity through interaction with PI4KB and neuronal calcium sensor-1 or calneuron-1. Both APOLs are present in Golgi-derived autophagy-related protein 9A vesicles, which are involved in PI4KB trafficking. Like APOL3-KO, APOL1Δ induces PI4KB dissociation from APOL3, linked to reduction of mitophagy flux and production of mitochondrial reactive oxygen species. APOL1 and APOL3, respectively, can interact with the mitophagy receptor prohibitin-2 and the mitophagosome membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). While APOL1 conditions PI4KB and APOL3 involvement in mitochondrion fission and mitophagy, APOL3-VAMP8 interaction promotes fusion between mitophagosomal and endolysosomal membranes. We propose that APOL3 controls mitochondrial membrane dynamics through interactions with the fission factor PI4KB and the fusion factor VAMP8.
Collapse
Affiliation(s)
- Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Paul Heo
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France; Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Maria Kløjgaard Skytthe
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | | | - Frédéric Pincet
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
15
|
Zhu JY, Lee JG, Fu Y, van de Leemput J, Ray PE, Han Z. APOL1-G2 accelerates nephrocyte cell death by inhibiting the autophagy pathway. Dis Model Mech 2023; 16:dmm050223. [PMID: 37969018 PMCID: PMC10765414 DOI: 10.1242/dmm.050223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/30/2023] [Indexed: 11/17/2023] Open
Abstract
People of African ancestry who carry the APOL1 risk alleles G1 or G2 are at high risk of developing kidney diseases through not fully understood mechanisms that impair the function of podocytes. It is also not clear whether the APOL1-G1 and APOL1-G2 risk alleles affect these cells through similar mechanisms. Previously, we have developed transgenic Drosophila melanogaster lines expressing either the human APOL1 reference allele (G0) or APOL1-G1 specifically in nephrocytes, the cells homologous to mammalian podocytes. We have found that nephrocytes that expressed the APOL1-G1 risk allele display accelerated cell death, in a manner similar to that of cultured human podocytes and APOL1 transgenic mouse models. Here, to compare how the APOL1-G1 and APOL1-G2 risk alleles affect the structure and function of nephrocytes in vivo, we generated nephrocyte-specific transgenic flies that either expressed the APOL1-G2 or both G1 and G2 (G1G2) risk alleles on the same allele. We found that APOL1-G2- and APOL1-G1G2-expressing nephrocytes developed more severe changes in autophagic pathways, acidification of organelles and the structure of the slit diaphragm, compared to G1-expressing nephrocytes, leading to their premature death. We conclude that both risk alleles affect similar key cell trafficking pathways, leading to reduced autophagy and suggesting new therapeutic targets to prevent APOL1 kidney diseases.
Collapse
Affiliation(s)
- Jun-yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jin-Gu Lee
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yulong Fu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Patricio E. Ray
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA 22903, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Wang XK, Guo YX, Wang M, Zhang XD, Liu ZY, Wang MS, Luo K, Huang S, Li RF. Identification and validation of candidate clinical signatures of apolipoprotein L isoforms in hepatocellular carcinoma. Sci Rep 2023; 13:20969. [PMID: 38017264 PMCID: PMC10684526 DOI: 10.1038/s41598-023-48366-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/25/2023] [Indexed: 11/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy worldwide with an increasing number of new cases each year. Apolipoprotein (APOL) isoforms have been explored for their associations with HCC.The GSE14520 cohort was used for training data; The Cancer Genome Atlas (TCGA) database was used for validated data. Diagnostic, prognostic significance and mechanisms were explored using these cohorts. Risk score models and nomograms were constructed using prognosis-related isoforms and clinical factors for survival prediction. Oncomine and HCCDB databases were further used for validation of diagnostic, prognostic significance. APOL1, 3, and 6 were differentially expressed in two cohorts (all P ≤ 0.05). APOL1 and APOL6 had diagnostic capacity whereas APOL3 and APOL6 had prognostic capacity in two cohorts (areas under curves [AUCs] > 0.7, P ≤ 0.05). Mechanism studies demonstrated that APOL3 and APOL6 might be involved in humoral chemokine signaling pathways (all P ≤ 0.05). Risk score models and nomograms were constructed and validated for survival prediction of HCC. Moreover, diagnostic values of APOL1 and weak APOL6 were validated in Oncomine database (AUC > 0.700, 0.694); prognostic values of APOL3 and APOL6 were validated in HCCDB database (all P < 0.05). Differentially expressed APOL1 and APOL6 might be diagnostic biomarkers; APOL3 and APOL6 might be prognostic biomarkers of RFS and OS for HCC via chemokine signaling pathways.
Collapse
Affiliation(s)
- Xiang-Kun Wang
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Yu-Xiang Guo
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Miao Wang
- Department of Gastrointestinal Oncology, Nanyang Second General Hospital, Nanyang, 473009, Henan Province, People's Republic of China
| | - Xu-Dong Zhang
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Zhong-Yuan Liu
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Mao-Sen Wang
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Kai Luo
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Shuai Huang
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Ren-Feng Li
- Departments of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
17
|
Lee JG, Fu Y, Zhu JY, Wen P, van de Leemput J, Ray PE, Han Z. A SNARE protective pool antagonizes APOL1 renal toxicity in Drosophila nephrocytes. Cell Biosci 2023; 13:199. [PMID: 37925499 PMCID: PMC10625211 DOI: 10.1186/s13578-023-01147-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/12/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND People of Sub-Saharan African ancestry are at higher risk of developing chronic kidney disease (CKD), attributed to the Apolipoprotein L1 (APOL1) gene risk alleles (RA) G1 and G2. The underlying mechanisms by which the APOL1-RA precipitate CKD remain elusive, hindering the development of potential treatments. RESULTS Using a Drosophila genetic modifier screen, we found that SNARE proteins (Syx7, Ykt6, and Syb) play an important role in preventing APOL1 cytotoxicity. Reducing the expression of these SNARE proteins significantly increased APOL1 cytotoxicity in fly nephrocytes, the equivalent of mammalian podocytes, whereas overexpression of Syx7, Ykt6, or Syb attenuated their toxicity in nephrocytes. These SNARE proteins bound to APOL1-G0 with higher affinity than APOL1-G1/G2, and attenuated APOL1-G0 cytotoxicity to a greater extent than either APOL1-RA. CONCLUSIONS Using a Drosophila screen, we identified SNARE proteins (Syx7, Ykt6, and Syb) as antagonists of APOL1-induced cytotoxicity by directly binding APOL1. These data uncovered a new potential protective role for certain SNARE proteins in the pathogenesis of APOL1-CKD and provide novel therapeutic targets for APOL1-associated nephropathies.
Collapse
Affiliation(s)
- Jin-Gu Lee
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yulong Fu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL, 35249, USA
| | - Jun-Yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Pei Wen
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Patricio E Ray
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, Charlottesville, VA, 22908, USA.
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine (UMSOM), 670 West Baltimore Street, 4052 HSFIII, Baltimore, MD, 21201, USA.
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
Dogan M, Watkins C, Ingram H, Moore N, Rucker GM, Gower EG, Eason JD, Bhalla A, Talwar M, Nezakatgoo N, Eymard C, Helmick R, Vanatta J, Bajwa A, Kuscu C, Kuscu C. Unveiling APOL1 Haplotypes: A Novel Classification Through Probe-Independent Quantitative Real-Time PCR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562539. [PMID: 37905084 PMCID: PMC10614821 DOI: 10.1101/2023.10.16.562539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Introduction Apolipoprotein-L1 (APOL1) is a primate-specific protein component of high- density lipoprotein (HDL). Two variants of APOL1 (G1 and G2), provide resistance to parasitic infections in African Americans but are also implicated in kidney-related diseases and transplant outcomes in recipients. This study aims to identify these risk variants using a novel probe- independent quantitative real-time PCR method in a high African American recipient cohort. Additionally, it aims to develop a new stratification approach based on haplotype-centric model. Methods Genomic DNA was extracted from recipient PBMCs using SDS lysis buffer and proteinase K. Quantitative PCR assay with modified forward primers and a common reverse primer enabled us to identify single nucleotide polymorphisms (SNPs) and the 6-bp deletion quantitatively. Additionally, we used sanger sequencing to verify our QPCR findings. Results Our novel probe-independent qPCR effectively distinguished homozygous wild-type, heterozygous SNPs/deletion, and homozygous SNPs/deletion, with at least 4-fold differences. High prevalence of APOL1 variants was observed (18% two-risk alleles, 34% one-risk allele) in our recipient cohort. Intriguingly, up to 12-month follow-up revealed no significant impact of recipient APOL1 variants on transplant outcomes. Ongoing research will encompass more time points and a larger patient cohort, allowing a comprehensive evaluation of G1/G2 variant subgroups categorized by new haplotype scores, enriching our understanding. Conclusions Our cost-effective and rapid qPCR technique facilitates APOL1 genotyping within hours. Prospective and retrospective studies will enable comparisons with long-term allograft rejection, potentially predicting early/late-stage transplant outcomes based on haplotype evaluation in this diverse group of kidney transplant recipients.
Collapse
|
19
|
Shen J, Zhao S, Peng M, Li Y, Zhang L, Li X, Hu Y, Wu M, Xiang S, Wu X, Liu J, Zhang B, Chen Z, Lin D, Liu H, Tang W, Chen J, Sun X, Liao Q, Hide G, Zhou Z, Lun ZR, Wu Z. Macrophage-mediated trogocytosis contributes to destroying human schistosomes in a non-susceptible rodent host, Microtus fortis. Cell Discov 2023; 9:101. [PMID: 37794085 PMCID: PMC10550985 DOI: 10.1038/s41421-023-00603-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Schistosoma parasites, causing schistosomiasis, exhibit typical host specificity in host preference. Many mammals, including humans, are susceptible to infection, while the widely distributed rodent, Microtus fortis, exhibits natural anti-schistosome characteristics. The mechanisms of host susceptibility remain poorly understood. Comparison of schistosome infection in M. fortis with the infection in laboratory mice (highly sensitive to infection) offers a good model system to investigate these mechanisms and to gain an insight into host specificity. In this study, we showed that large numbers of leukocytes attach to the surface of human schistosomes in M. fortis but not in mice. Single-cell RNA-sequencing analyses revealed that macrophages might be involved in the cell adhesion, and we further demonstrated that M. fortis macrophages could be mediated to attach and kill schistosomula with dependence on Complement component 3 (C3) and Complement receptor 3 (CR3). Importantly, we provided direct evidence that M. fortis macrophages could destroy schistosomula by trogocytosis, a previously undescribed mode for killing helminths. This process was regulated by Ca2+/NFAT signaling. These findings not only elucidate a novel anti-schistosome mechanism in M. fortis but also provide a better understanding of host parasite interactions, host specificity and the potential generation of novel strategies for schistosomiasis control.
Collapse
Affiliation(s)
- Jia Shen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| | - Siyu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mei Peng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Yanguo Li
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lichao Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center, Organ Transplantation Institute, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yunyi Hu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mingrou Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Suoyu Xiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoying Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahua Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Beibei Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Zebin Chen
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Huanyao Liu
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyan Tang
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Chen
- Department of Immunology, Center for Precision Medicine and Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Qi Liao
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK
| | - Zhijun Zhou
- Department of Laboratory Animals, Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, China.
| | - Zhao-Rong Lun
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK.
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Lorentzen J, Olesen HG, Hansen AG, Thiel S, Birkelund S, Andersen CBF, Andersen GR. Trypanosoma brucei Invariant Surface gp65 Inhibits the Alternative Pathway of Complement by Accelerating C3b Degradation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:862-873. [PMID: 37466368 DOI: 10.4049/jimmunol.2300128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023]
Abstract
Trypanosomes are known to activate the complement system on their surface, but they control the cascade in a manner such that the cascade does not progress into the terminal pathway. It was recently reported that the invariant surface glycoprotein ISG65 from Trypanosoma brucei interacts reversibly with complement C3 and its degradation products, but the molecular mechanism by which ISG65 interferes with complement activation remains unknown. In this study, we show that ISG65 does not interfere directly with the assembly or activity of the two C3 convertases. However, ISG65 acts as a potent inhibitor of C3 deposition through the alternative pathway in human and murine serum. Degradation assays demonstrate that ISG65 stimulates the C3b to iC3b converting activity of complement factor I in the presence of the cofactors factor H or complement receptor 1. A structure-based model suggests that ISG65 promotes a C3b conformation susceptible to degradation or directly bridges factor I and C3b without contact with the cofactor. In addition, ISG65 is observed to form a stable ternary complex with the ligand binding domain of complement receptor 3 and iC3b. Our data suggest that ISG65 supports trypanosome complement evasion by accelerating the conversion of C3b to iC3b through a unique mechanism.
Collapse
Affiliation(s)
- Josefine Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Heidi G Olesen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | | | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
21
|
Oula JO, Mose JM, Waiganjo NN, Chepukosi KW, Mitalo NS, Isaac AO, Nyariki JN. Vitamin B12 blocked Trypanosoma brucei rhodesiense-driven disruption of the blood brain barrier, and normalized nitric oxide and malondialdehyde levels in a mouse model. Parasitol Int 2023; 96:102775. [PMID: 37390918 DOI: 10.1016/j.parint.2023.102775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Infection with Trypanosoma brucei rhodesiense (T.b.r) causes acute Human African Trypanosomiasis (HAT) in Africa. This study determined the effect of vitamin B12 on T.b.r -driven pathological events in a mouse model. Mice were randomly assigned into four groups; group one was the control. Group two was infected with T.b.r; group three was supplemented with 8 mg/kg vitamin B12 for two weeks; before infection with T.b.r. For group four, administration of vitamin B12 was started from the 4th days post-infection with T.b.r. At 40 days post-infection, the mice were sacrificed to obtain blood, tissues, and organs for various analyses. The results showed that vitamin B12 administration enhanced the survival rate of T.b.r infected mice, and prevented T.b.r-induced disruption of the blood-brain barrier and decline in neurological performance. Notably, T.b.r-induced hematological alteration leading to anaemia, leukocytosis and dyslipidemia was alleviated by vitamin B12. T.b.r-induced elevation of the liver alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and total bilirubin as well as the kidney damage markers urea, uric acid and creatinine were attenuated by vitamin B12. Vitamin B12 blocked T.b.r-driven rise in TNF-α and IFN-γ, nitric oxide and malondialdehyde. T.b.r-induced depletion of GSH levels were attenuated in the presence of vitamin B12 in the brain, spleen and liver tissues; a clear indication of the antioxidant activity of vitamin B12. In conclusion, treatment with vitamin B12 potentially protects against various pathological events associated with severe late-stage HAT and presents a great opportunity for further scrutiny to develop an adjunct therapy for severe late-stage HAT.
Collapse
Affiliation(s)
- James O Oula
- Department of Biomedical Science & Technology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - John Mokua Mose
- Department of Biomedical Science & Technology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - Naomi N Waiganjo
- Department of Biomedical Science & Technology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - Kennedy W Chepukosi
- Department of Biochemistry and Biotechnology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - Nancy S Mitalo
- Department of Biomedical Science & Technology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - Alfred Orina Isaac
- Department of Pharmaceutical Sciences and Technology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya
| | - James Nyabuga Nyariki
- Department of Biochemistry and Biotechnology, Technical University of Kenya, P. O. Box 52428, 00200 Nairobi, Kenya.
| |
Collapse
|
22
|
Carracedo M, Ericson E, Ågren R, Forslöw A, Madeyski-Bengtson K, Svensson A, Riddle R, Christoffersson J, González-King Garibotti H, Lazovic B, Hicks R, Buvall L, Fornoni A, Greasley PJ, Lal M. APOL1 promotes endothelial cell activation beyond the glomerulus. iScience 2023; 26:106830. [PMID: 37250770 PMCID: PMC10209455 DOI: 10.1016/j.isci.2023.106830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk genotypes are associated with increased risk of chronic kidney disease (CKD) in people of West African ancestry. Given the importance of endothelial cells (ECs) in CKD, we hypothesized that APOL1 high-risk genotypes may contribute to disease via EC-intrinsic activation and dysfunction. Single cell RNA sequencing (scRNA-seq) analysis of the Kidney Precision Medicine Project dataset revealed APOL1 expression in ECs from various renal vascular compartments. Utilizing two public transcriptomic datasets of kidney tissue from African Americans with CKD and a dataset of APOL1-expressing transgenic mice, we identified an EC activation signature; specifically, increased intercellular adhesion molecule 1 (ICAM-1) expression and enrichment in leukocyte migration pathways. In vitro, APOL1 expression in ECs derived from genetically modified human induced pluripotent stem cells and glomerular ECs triggered changes in ICAM-1 and platelet endothelial cell adhesion molecule 1 (PECAM-1) leading to an increase in monocyte attachment. Overall, our data suggest the involvement of APOL1 as an inducer of EC activation in multiple renal vascular beds with potential effects beyond the glomerular vasculature.
Collapse
Affiliation(s)
- Miguel Carracedo
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elke Ericson
- Genome Engineering, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Ågren
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Forslöw
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Katja Madeyski-Bengtson
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Svensson
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rebecca Riddle
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Jonas Christoffersson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bojana Lazovic
- Genome Engineering, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Lisa Buvall
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Peter J. Greasley
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mark Lal
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
23
|
Sülzen H, Began J, Dhillon A, Kereïche S, Pompach P, Votrubova J, Zahedifard F, Šubrtova A, Šafner M, Hubalek M, Thompson M, Zoltner M, Zoll S. Cryo-EM structures of Trypanosoma brucei gambiense ISG65 with human complement C3 and C3b and their roles in alternative pathway restriction. Nat Commun 2023; 14:2403. [PMID: 37105991 PMCID: PMC10140031 DOI: 10.1038/s41467-023-37988-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
African Trypanosomes have developed elaborate mechanisms to escape the adaptive immune response, but little is known about complement evasion particularly at the early stage of infection. Here we show that ISG65 of the human-infective parasite Trypanosoma brucei gambiense is a receptor for human complement factor C3 and its activation fragments and that it takes over a role in selective inhibition of the alternative pathway C5 convertase and thus abrogation of the terminal pathway. No deposition of C4b, as part of the classical and lectin pathway convertases, was detected on trypanosomes. We present the cryo-electron microscopy (EM) structures of native C3 and C3b in complex with ISG65 which reveal a set of modes of complement interaction. Based on these findings, we propose a model for receptor-ligand interactions as they occur at the plasma membrane of blood-stage trypanosomes and may facilitate innate immune escape of the parasite.
Collapse
Affiliation(s)
- Hagen Sülzen
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
- Faculty of Science, Charles University, Albertov 6, 12800, Prague 2, Czech Republic
| | - Jakub Began
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
- Department of Immunobiology, University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Arun Dhillon
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
| | - Sami Kereïche
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Albertov 4, 12800, Prague, Czech Republic
| | - Petr Pompach
- Institute of Biotechnology of the Czech Academy of Sciences, 25250, Vestec, Czech Republic
| | - Jitka Votrubova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
| | - Farnaz Zahedifard
- Department of Parasitology, Faculty of Science, Charles University Prague, Biocev, 25250, Vestec, Czech Republic
| | - Adriana Šubrtova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
| | - Marie Šafner
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
| | - Martin Hubalek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
| | - Maaike Thompson
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic
- University of Antwerp, Antwerp, Belgium
- Agidens, Industrial Machinery Manufacturing, Zwijndrecht, Antwerp, Belgium
| | - Martin Zoltner
- Department of Parasitology, Faculty of Science, Charles University Prague, Biocev, 25250, Vestec, Czech Republic
| | - Sebastian Zoll
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000, Prague, Czech Republic.
| |
Collapse
|
24
|
Marcos LA, Vorkas CK, Mann I, Garry E, Lamba P, Pham SK, Spector R, Papamanoli A, Krivacsy S, Lum M, Zahra A, Hou W, Spitzer ED. Reduced Cholesterol Levels during Acute Human Babesiosis. Pathogens 2023; 12:pathogens12040613. [PMID: 37111499 PMCID: PMC10144092 DOI: 10.3390/pathogens12040613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Babesiosis, an intra-erythrocytic protozoan disease, is an emerging zoonotic parasitic disease worldwide. Cholesterol levels are correlated with severe infections, such as sepsis and COVID-19, and anecdotal reports suggest that high-density lipoprotein (HDL) cholesterol declines during acute babesiosis. Our aim was to describe the cholesterol levels in patients with acute babesiosis diagnosed in an endemic area in New York, hypothesizing that HDL levels correlate with the severity of infection. METHODS We reviewed the medical records of adult patients with babesiosis diagnosed by identification of Babesia parasites on a thin blood smear and confirmed by polymerase chain reaction from 2013 to 2018, who also had available a lipid profile drawn at the time of clinical presentation. Additional lipid profile levels were considered as "baseline" if they were drawn within 2 months before or after the infection as part of routine care. RESULTS A total of 39 patients with babesiosis had a lipid profile drawn on presentation. The patients were divided into two groups for comparison based on the treating physician's clinical decision: 33 patients who were admitted to the hospital and 8 patients who were evaluated as outpatients. A history of hypertension was more common in admitted patients (37% vs. 17%, p = 0.02). The median levels of low-density lipoprotein (LDL) and HDL were significantly reduced in admitted patients compared to non-admitted patients (46 vs. 76 mg/dL, p = 0.04; and 9 vs. 28.5 mg/dL, p = 0.03, respectively). In addition, LDL and HDL levels returned to baseline values following resolution of acute babesiosis. CONCLUSION LDL and HDL levels are significantly reduced during acute babesiosis, suggesting that cholesterol depletion may predict disease severity. Pathogen and host factors may contribute to a reduction in serum cholesterol levels during acute babesiosis.
Collapse
Affiliation(s)
- Luis A Marcos
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Charles Kyriakos Vorkas
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Inderjit Mann
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Evan Garry
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Pooja Lamba
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sophia K Pham
- Stony Brook Southampton Hospital, Southampton, NY 11968, USA
| | - Rachel Spector
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Aikaterini Papamanoli
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sara Krivacsy
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael Lum
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Aleena Zahra
- Division of Infectious Diseases, Department of Internal Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Wei Hou
- Division of Epidemiology and Biostatistics, Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Eric D Spitzer
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
25
|
Makarov A, Began J, Mautone IC, Pinto E, Ferguson L, Zoltner M, Zoll S, Field MC. The role of invariant surface glycoprotein 75 in xenobiotic acquisition by African trypanosomes. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:18-35. [PMID: 36789350 PMCID: PMC9896412 DOI: 10.15698/mic2023.02.790] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 02/05/2023]
Abstract
The surface proteins of parasitic protozoa mediate functions essential to survival within a host, including nutrient accumulation, environmental sensing and immune evasion. Several receptors involved in nutrient uptake and defence from the innate immune response have been described in African trypanosomes and, together with antigenic variation, contribute towards persistence within vertebrate hosts. Significantly, a superfamily of invariant surface glycoproteins (ISGs) populates the trypanosome surface, one of which, ISG75, is implicated in uptake of the century-old drug suramin. By CRISPR/Cas9 knockout and biophysical analysis, we show here that ISG75 directly binds suramin and mediates uptake of additional naphthol-related compounds, making ISG75 a conduit for entry of at least one structural class of trypanocidal compounds. However, ISG75 null cells present only modest attenuation of suramin sensitivity, have unaltered viability in vivo and in vitro and no alteration to suramin-invoked proteome responses. While ISG75 is demonstrated as a valid suramin cell entry pathway, we suggest the presence of additional mechanisms for suramin accumulation, further demonstrating the complexity of trypanosomatid drug interactions and potential for evolution of resistance.
Collapse
Affiliation(s)
- Alexandr Makarov
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Jakub Began
- Laboratory of Structural Parasitology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague 6, Czech Republic
| | - Ileana Corvo Mautone
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Laboratorio de Moléculas Bioactivas, Departamento de Ciencias Biológicas, Universidad de la República, Paysandú 60000, Uruguay
| | - Erika Pinto
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Liam Ferguson
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Martin Zoltner
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Charles University, Faculty of Science, Department of Parasitology, Vestec, Czech Republic
| | - Sebastian Zoll
- Laboratory of Structural Parasitology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague 6, Czech Republic
| | - Mark C. Field
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| |
Collapse
|
26
|
Abstract
African trypanosomes are bloodstream protozoan parasites that infect mammals including humans, where they cause sleeping sickness. Long-lasting infection is required to favor parasite transmission between hosts. Therefore, trypanosomes have developed strategies to continuously escape innate and adaptive responses of the immune system, while also preventing premature death of the host. The pathology linked to infection mainly results from inflammation and includes anemia and brain dysfunction in addition to loss of specificity and memory of the antibody response. The serum of humans contains an efficient trypanolytic factor, the membrane pore-forming protein apolipoprotein L1 (APOL1). In the two human-infective trypanosomes, specific parasite resistance factors inhibit APOL1 activity. In turn, many African individuals express APOL1 variants that counteract these resistance factors, enabling them to avoid sleeping sickness. However, these variants are associated with chronic kidney disease, particularly in the context of virus-induced inflammation such as coronavirus disease 2019. Vaccination perspectives are discussed.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, Université Libre de Bruxelles, Gosselies, Belgium;
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium;
| | - Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; .,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
27
|
Heme-deficient metabolism and impaired cellular differentiation as an evolutionary trade-off for human infectivity in Trypanosoma brucei gambiense. Nat Commun 2022; 13:7075. [PMID: 36400774 PMCID: PMC9674590 DOI: 10.1038/s41467-022-34501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Resistance to African trypanosomes in humans relies in part on the high affinity targeting of a trypanosome lytic factor 1 (TLF1) to a trypanosome haptoglobin-hemoglobin receptor (HpHbR). While TLF1 avoidance by the inactivation of HpHbR contributes to Trypanosoma brucei gambiense human infectivity, the evolutionary trade-off of this adaptation is unknown, as the physiological function of the receptor remains to be elucidated. Here we show that uptake of hemoglobin via HpHbR constitutes the sole heme import pathway in the trypanosome bloodstream stage. T. b. gambiense strains carrying the inactivating mutation in HpHbR, as well as genetically engineered T. b. brucei HpHbR knock-out lines show only trace levels of intracellular heme and lack hemoprotein-based enzymatic activities, thereby providing an uncommon example of aerobic parasitic proliferation in the absence of heme. We further show that HpHbR facilitates the developmental progression from proliferating long slender forms to cell cycle-arrested stumpy forms in T. b. brucei. Accordingly, T. b. gambiense was found to be poorly competent for slender-to-stumpy differentiation unless a functional HpHbR receptor derived from T. b. brucei was genetically restored. Altogether, we identify heme-deficient metabolism and disrupted cellular differentiation as two distinct HpHbR-dependent evolutionary trade-offs for T. b. gambiense human infectivity.
Collapse
|
28
|
Abdu A, Duarte R, Dickens C, Dix-Peek T, Bala SM, Ademola B, Naicker S. High risk APOL1 genotypes and kidney disease among treatment naïve HIV patients at Kano, Nigeria. PLoS One 2022; 17:e0275949. [PMID: 36227935 PMCID: PMC9560498 DOI: 10.1371/journal.pone.0275949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/26/2022] [Indexed: 11/05/2022] Open
Abstract
Introduction Racial disparities are known in the occurrence of kidney disease with excess risks found among people of African descent. Apolipoprotein L1 (APOL1) gene variants G1 and G2 are associated with kidney disease among HIV infected individuals of African descent in the USA as well as among black population in South Africa. We set out to investigate the prevalence of these high-risk variants and their effects on kidney disease among HIV infected patients in Northern Nigeria with hitherto limited information despite earlier reports of high population frequencies of these alleles from the Southern part of the country. Methods DNA samples obtained from the whole blood of 142 participants were genotyped for APOL1 G1 and G2 variants after initial baseline investigations including assessment of kidney function. Participants comprised 50 HIV positive patients with no evidence of kidney disease, 52 HIV negative individuals with no kidney disease and 40 HIV positive patients with chronic kidney disease (CKD) evidenced by persistent proteinuria and/or reduced eGFR, who also had a kidney biopsy. All the HIV positive patients were newly diagnosed and treatment naïve. Results The distribution of the APOL1 genotypes among the study participants revealed that 24.6% had a G1 risk allele and 19.0% a G2. The frequency of the High Risk Genotype (HRG) was 12.5% among those with CKD compared to 5.8% in the HIV negative group and zero in the HIV positive no CKD group. Having the HRG was associated with a higher odds for developing HIV Associated Nephropathy (HIVAN) (2 vs 0 risk alleles: OR 10.83, 95% CI 1.38–84.52; P = 0.023; 2 vs 0 or 1 risk alleles: OR 5.5, 95% CI 0.83–36.29; P = 0.07). The HRG was also associated with higher odds for Focal Segmental Glomerulosclerosis (FSGS) (2 vs 0 risk alleles: OR 13.0, 95% CI 2.06–81.91; P = 0.006 and 2 vs 0 or 1 risk alleles: OR 9.0, 95%CI 1.62–50.12; P = 0.01) when compared to the control group. Conclusion This study showed a high population frequency of the individual risk alleles of the APOL1 gene with higher frequencies noted among HIV positive patients with kidney disease. There is high association with the presence of kidney disease and especially FSGS and HIVAN among treatment naive HIV patients carrying two copies of the HRG.
Collapse
Affiliation(s)
- Aliyu Abdu
- Department of Medicine Aminu Kano Teaching Hospital/ Bayero University Kano, Kano, Nigeria
- * E-mail:
| | - Raquel Duarte
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline Dickens
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Therese Dix-Peek
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sunusi M. Bala
- Department of Medicine, M.A. Wase Teaching Hospital, Kano, Nigeria
| | - Babatunde Ademola
- Department of Medicine Aminu Kano Teaching Hospital/ Bayero University Kano, Kano, Nigeria
| | - Saraladevi Naicker
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
29
|
Alladagbin DJ, da Silva CGR, Silva LK, Dos-Santos WL, de Sá Oliveira GG. APOL1 genotyping via buccal mucosa cell samples to establish risk of kidney disease. BMC Nephrol 2022; 23:329. [PMID: 36217118 PMCID: PMC9549854 DOI: 10.1186/s12882-022-02954-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/18/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022] Open
Abstract
Two alleles (G1 and G2) of the apolipoprotein 1 gene (APOL1) predispose people of African descent to developing or accelerating the course of certain types of kidney disease. Population studies to determine the frequency of the G1 and G2 alleles are important to inform resource allocation by public health authorities. Traditionally, APOL1 genotyping is carried out in blood samples. However, sample collection, transport, and storage is cumbersome. Here we compared APOL1 genotyping in blood and buccal mucosa cell samples obtained from 23 individuals. Alleles G0 (wild), G1, and G2, as well as genotypes G0/G0, G0/G1, G1/G1, G0/G2, G1/G2, and G2/G2 were detected using both blood and buccal mucosa cells with 100% coincidence. Our data indicate that buccal mucosa cell samples may represent a suitable alternative to blood samples for APOL1 genotyping in the field.
Collapse
Affiliation(s)
- Dona Jeanne Alladagbin
- Fundação Oswaldo Cruz, Laboratory of Molecular and Strutural Pathology (LAPEM), Gonçalo Moniz Institute, FIOCRUZ-BA, Rua Waldemar Falcão, no. 121, CEP 40.296-710, Salvador, Brazil
| | - Carlos Gustavo Regis da Silva
- Fundação Oswaldo Cruz, Laboratory of Molecular and Strutural Pathology (LAPEM), Gonçalo Moniz Institute, FIOCRUZ-BA, Rua Waldemar Falcão, no. 121, CEP 40.296-710, Salvador, Brazil
| | - Luciano Kalabric Silva
- Fundação Oswaldo Cruz, Laboratory of Molecular and Strutural Pathology (LAPEM), Gonçalo Moniz Institute, FIOCRUZ-BA, Rua Waldemar Falcão, no. 121, CEP 40.296-710, Salvador, Brazil
| | - Washington Lc Dos-Santos
- Fundação Oswaldo Cruz, Laboratory of Molecular and Strutural Pathology (LAPEM), Gonçalo Moniz Institute, FIOCRUZ-BA, Rua Waldemar Falcão, no. 121, CEP 40.296-710, Salvador, Brazil
| | - Geraldo Gileno de Sá Oliveira
- Fundação Oswaldo Cruz, Laboratory of Molecular and Strutural Pathology (LAPEM), Gonçalo Moniz Institute, FIOCRUZ-BA, Rua Waldemar Falcão, no. 121, CEP 40.296-710, Salvador, Brazil.
| |
Collapse
|
30
|
Skytthe MK, Sørensen AL, Hennig D, Sandberg MB, Rasmussen LM, Højrup P, Møller HJ, Skjødt K, Moestrup SK, Graversen JH. Haptoglobin-related protein in human plasma correlates to haptoglobin concentrations and phenotypes. Scandinavian Journal of Clinical and Laboratory Investigation 2022; 82:461-466. [PMID: 36129375 DOI: 10.1080/00365513.2022.2122076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Haptoglobin-related protein (Hpr) is a plasma protein with high sequence similarity to haptoglobin (Hp). Like Hp, Hpr also binds hemoglobin (Hb) with high affinity, but it does not bind to the Hb-Hp receptor CD163 on macrophages. The Hpr concentration is markedly lower than Hp in plasma and its regulation is not understood. In the present study, we have developed non-crossreactive antibodies to Hpr to analyze the Hpr concentration in 112 plasma samples from anonymized individuals and compared it to Hp. The results show that plasma Hpr correlated with Hp concentrations (rho = 0.46, p = .0001). Hpr accounts for on average 0.35% of the Hp/Hpr pool but up to 29% at low Hp levels. Furthermore, the Hpr concentrations were significantly lower in individuals with the Hp2-2 phenotype compared to those with the Hp2-1 or Hp1-1 phenotypes. Experimental binding analysis did not provide evidence that Hpr associates with Hp and in this way is removed via CD163. In conclusion, the Hpr concentration correlates to Hp concentrations and Hp-phenotypes by yet unknown mechanisms independent of CD163-mediated removal of Hb-Hp complexes.
Collapse
Affiliation(s)
- Maria Kløjgaard Skytthe
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark
| | - Anna Lahn Sørensen
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark
| | - Dorle Hennig
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark
| | - Maria Boysen Sandberg
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense C, Denmark
| | - Lars Melholt Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense C, Denmark
| | - Peter Højrup
- Department of Biochemistry and molecular biology, University of Southern Denmark, Odense M, Denmark
| | - Holger J Møller
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark.,Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus N, Denmark
| | - Karsten Skjødt
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark
| | - Søren Kragh Moestrup
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark.,Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus N, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jonas Heilskov Graversen
- Department of Molecular Medicine-Cancer and Inflammation Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
31
|
Ilori T, Watanabe A, Ng KH, Solarin A, Sinha A, Gbadegesin R. Genetics of Chronic Kidney Disease in Low-Resource Settings. Semin Nephrol 2022; 42:151314. [PMID: 36801667 PMCID: PMC10272019 DOI: 10.1016/j.semnephrol.2023.151314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Advances in kidney genomics in the past 20 years has opened the door for more precise diagnosis of kidney disease and identification of new and specific therapeutic agents. Despite these advances, an imbalance exists between low-resource and affluent regions of the world. Individuals of European ancestry from the United States, United Kingdom, and Iceland account for 16% of the world's population, but represent more than 80% of all genome-wide association studies. South Asia, Southeast Asia, Latin America, and Africa together account for 57% of the world population but less than 5% of genome-wide association studies. Implications of this difference include limitations in new variant discovery, inaccurate interpretation of the effect of genetic variants in non-European populations, and unequal access to genomic testing and novel therapies in resource-poor regions. It also further introduces ethical, legal, and social pitfalls, and ultimately may propagate global health inequities. Ongoing efforts to reduce the imbalance in low-resource regions include funding and capacity building, population-based genome sequencing, population-based genome registries, and genetic research networks. More funding, training, and capacity building for infrastructure and expertise is needed in resource-poor regions. Focusing on this will ensure multiple-fold returns on investments in genomic research and technology.
Collapse
Affiliation(s)
- Titilayo Ilori
- Division of Nephrology, Boston University School of Medicine, Boston, MA
| | - Andreia Watanabe
- Division of Molecular Medicine, Department of Pediatrics, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Kar-Hui Ng
- Department of Pediatrics, Yong Loo Lin School of Medicine, Singapore
| | - Adaobi Solarin
- Department of Pediatrics and Child Health, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Aditi Sinha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rasheed Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
32
|
Sengupta PP, Jacob SS, Chandu AGS, Das S. Silent Trypanosoma evansi infection in humans from India revealed by serological and molecular surveys, and characterized by variable surface glycoprotein gene sequences. Acta Trop 2022; 229:106369. [PMID: 35157840 DOI: 10.1016/j.actatropica.2022.106369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The importance of emerging atypical human trypanosomosis is gaining momentum due to increasing detection and its possible impact on human health. A cross sectional study of atypical human trypanosomosis due to Trypanosoma evansi was carried out in Kolkata and Canning area of West Bengal state of India where previously a death was reported. METHODS In this study blood and serum samples from 173 individuals were collected during August to December 2014. To check the presence of antibodies against T. evansi, card agglutination test and for the presence of T. evansi specific DNA, PCR were conducted. RESULTS T. evansi infection was identified in 5.2% (9/173) human blood samples by CATT serological test (Card agglutination test for trypanosomosis). PCR targeting VSG gene sequences suggested active T. evansi infection in 2.89% (5/173). VSG gene sequences herein determined for five isolates from human cases shared high similarity (89.4-100%). Phylogenetic inference clustered the human isolates with other isolates from different host species from India and other countries, forming a clade exclusive of Indian isolates (84.0 to 100% sequence similarity). CONCLUSION First report of symptomless human T. evansi infection detected by combined serological and PCR assays. First phylogenetic analysis of VSG gene sequences including human isolates of T. evansi in which Indian isolates of T. evansi from human and other hosts clustered in a single clade.
Collapse
Affiliation(s)
- Pinaki Prasad Sengupta
- ICAR- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru - 560064, Karnataka, India
| | - Siju Susan Jacob
- ICAR- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru - 560064, Karnataka, India
| | - Atru Gnana Surya Chandu
- ICAR- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru - 560064, Karnataka, India
| | - Satadal Das
- Peerless Hospital and B. K. Roy Research Center, Kolkata - 700094, West Bengal, India.
| |
Collapse
|
33
|
Hubacek JA, Hruba P, Adamkova V, Pokorna E, Viklicky O. Apolipoprotein L1 variability is associated with increased risk of renal failure in the Czech population. Gene X 2022; 818:146248. [PMID: 35085711 DOI: 10.1016/j.gene.2022.146248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/23/2021] [Accepted: 01/21/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND With stage 5 chronic kidney disease (CKD5) more prevalent in the Czech Republic than in most European countries, genetic susceptibility is potentially implicated. METHODS In a group of 1489 CKD5 kidney transplantation patients (93% with complete clinical characteristics; mean age 52.0 years, 37% females) and 2559 healthy controls (mean age 49.0 years, 51% females), we examined the prevalence of six APOL1 SNPs (rs73885319, rs71785313, rs13056427, rs136147, rs10854688 and rs9610473) and one newly detected 55-nucleotide insertion/deletion polymorphism. RESULTS The rs73885319 and rs71785313 variants were monomorphic in the Czech Caucasian population. Genotype frequencies of the three SNPs examined (rs13056427, rs136147 and rs9610473) were almost identical in patients and controls (all P values were between 0.39 and 0.91). Minor homozygotes of rs10854688 were more common between the patients (13.2%) than in controls (10.7%) (OR [95% CI]; 1.32 [1.08-1.64]; P < 0.01). Prevalence of the newly detected 55-bp APOL1 deletion was significantly higher in CKD5 patients (3.0% vs. 1.7%; OR [95% CI]; 1.80 [1.16-2.80]; P < 0.01) compared to controls. Frequencies of some individual APOL1 haplotypes were borderline different between patients and controls. CONCLUSION We found an association between rs10854688 SNP within the APOL1 gene and end-stage renal disease in the Czech Caucasian population. Further independent studies are required before a conclusive association between the newly detected APOL1 insertion/deletion polymorphism and CKD5 can be confirmed.
Collapse
Affiliation(s)
- Jaroslav A Hubacek
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Petra Hruba
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vera Adamkova
- Department of Preventive Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Pokorna
- Transplantation Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondrej Viklicky
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Nephrology, Transplantation Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
34
|
Daneshpajouhnejad P, Kopp JB, Winkler CA, Rosenberg AZ. The evolving story of apolipoprotein L1 nephropathy: the end of the beginning. Nat Rev Nephrol 2022; 18:307-320. [PMID: 35217848 PMCID: PMC8877744 DOI: 10.1038/s41581-022-00538-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 01/13/2023]
Abstract
Genetic coding variants in APOL1, which encodes apolipoprotein L1 (APOL1), were identified in 2010 and are relatively common among individuals of sub-Saharan African ancestry. Approximately 13% of African Americans carry two APOL1 risk alleles. These variants, termed G1 and G2, are a frequent cause of kidney disease — termed APOL1 nephropathy — that typically manifests as focal segmental glomerulosclerosis and the clinical syndrome of hypertension and arterionephrosclerosis. Cell culture studies suggest that APOL1 variants cause cell dysfunction through several processes, including alterations in cation channel activity, inflammasome activation, increased endoplasmic reticulum stress, activation of protein kinase R, mitochondrial dysfunction and disruption of APOL1 ubiquitinylation. Risk of APOL1 nephropathy is mostly confined to individuals with two APOL1 risk variants. However, only a minority of individuals with two APOL1 risk alleles develop kidney disease, suggesting the need for a ‘second hit’. The best recognized factor responsible for this ‘second hit’ is a chronic viral infection, particularly HIV-1, resulting in interferon-mediated activation of the APOL1 promoter, although most individuals with APOL1 nephropathy do not have an obvious cofactor. Current therapies for APOL1 nephropathies are not adequate to halt progression of chronic kidney disease, and new targeted molecular therapies are in clinical trials. This Review summarizes current understanding of the role of APOL1 variants in kidney disease. The authors discuss the genetics, protein structure and biological functions of APOL1 variants and provide an overview of promising therapeutic strategies. In contrast to other APOL family members, which are primarily intracellular, APOL1 contains a unique secretory signal peptide, resulting in its secretion into plasma. APOL1 renal risk alleles provide protection from African human trypanosomiasis but are a risk factor for progressive kidney disease in those carrying two risk alleles. APOL1 risk allele frequency is ~35% in the African American population in the United States, with ~13% of individuals having two risk alleles; the highest allele frequencies are found in West African populations and their descendants. Cell and mouse models implicate endolysosomal and mitochondrial dysfunction, altered ion channel activity, altered autophagy, and activation of protein kinase R in the pathogenesis of APOL1-associated kidney disease; however, the relevance of these injury pathways to human disease has not been resolved. APOL1 kidney disease tends to be progressive, and current standard therapies are generally ineffective; targeted therapeutic strategies hold the most promise.
Collapse
Affiliation(s)
- Parnaz Daneshpajouhnejad
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Pennsylvania Hospital, Philadelphia, PA, USA
| | | | - Cheryl A Winkler
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
35
|
Desquesnes M, Gonzatti M, Sazmand A, Thévenon S, Bossard G, Boulangé A, Gimonneau G, Truc P, Herder S, Ravel S, Sereno D, Jamonneau V, Jittapalapong S, Jacquiet P, Solano P, Berthier D. A review on the diagnosis of animal trypanosomoses. Parasit Vectors 2022; 15:64. [PMID: 35183235 PMCID: PMC8858479 DOI: 10.1186/s13071-022-05190-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/01/2022] [Indexed: 01/07/2023] Open
Abstract
This review focuses on the most reliable and up-to-date methods for diagnosing trypanosomoses, a group of diseases of wild and domestic mammals, caused by trypanosomes, parasitic zooflagellate protozoans mainly transmitted by insects. In Africa, the Americas and Asia, these diseases, which in some cases affect humans, result in significant illness in animals and cause major economic losses in livestock. A number of pathogens are described in this review, including several Salivarian trypanosomes, such as Trypanosoma brucei sspp. (among which are the agents of sleeping sickness, the human African trypanosomiasis [HAT]), Trypanosoma congolense and Trypanosoma vivax (causing “Nagana” or animal African trypanosomosis [AAT]), Trypanosoma evansi (“Surra”) and Trypanosoma equiperdum (“Dourine”), and Trypanosoma cruzi, a Stercorarian trypanosome, etiological agent of the American trypanosomiasis (Chagas disease). Diagnostic methods for detecting zoonotic trypanosomes causing Chagas disease and HAT in animals, as well as a diagnostic method for detecting animal trypanosomes in humans (the so-called “atypical human infections by animal trypanosomes” [a-HT]), including T. evansi and Trypanosoma lewisi (a rat parasite), are also reviewed. Our goal is to present an integrated view of the various diagnostic methods and techniques, including those for: (i) parasite detection; (ii) DNA detection; and (iii) antibody detection. The discussion covers various other factors that need to be considered, such as the sensitivity and specificity of the various diagnostic methods, critical cross-reactions that may be expected among Trypanosomatidae, additional complementary information, such as clinical observations and epizootiological context, scale of study and logistic and cost constraints. The suitability of examining multiple specimens and samples using several techniques is discussed, as well as risks to technicians, in the context of specific geographical regions and settings. This overview also addresses the challenge of diagnosing mixed infections with different Trypanosoma species and/or kinetoplastid parasites. Improving and strengthening procedures for diagnosing animal trypanosomoses throughout the world will result in a better control of infections and will significantly impact on “One Health,” by advancing and preserving animal, human and environmental health.
Collapse
|
36
|
Pas-de-deux: African Plasmodium falciparum adaptations to sickle hemoglobin. Trends Parasitol 2022; 38:267-268. [DOI: 10.1016/j.pt.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/18/2022]
|
37
|
Comparative Analysis of the APOL1 Variants in the Genetic Landscape of Renal Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14030733. [PMID: 35159001 PMCID: PMC8833631 DOI: 10.3390/cancers14030733] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) occurs at higher frequency in individuals of African ancestry, with well-recorded documentation in this community. This is most prominent in the context of chronic kidney disease. In turn, many forms of progressive chronic kidney disease are more common in populations of Sub-Saharan African ancestry. This disparity has been attributed to well-defined allelic variants and has risen in the parental populations to high frequency under evolutionary pressure. Mechanisms of increased kidney disease risk and cell injury, causally associated with these APOL1 gene variants, have been extensively studied. Most studies have compared the effects of ectopic overexpression of the parental non-risk APOL1 with the mutated risk variants in cellular and organismal platforms. In the current study, we have used CRISPR/Cas9 genetic engineering to knock out or modify the sequence of endogenous APOL1 in RCC to mimic and examine the effects of these naturally occurring kidney disease risk allelic variants. Remarkably, these modifications to endogenous APOL1 genes in RCC resulted in a set of prominent effects on mitochondrial integrity and metabolic pathways and disrupted tumorigenesis. These findings both clarify pathways of cell injury of APOL1 risk variants in cells of kidney origin and motivate further studies to examine the potential central role of APOL1 in the pathogenesis of renal cell carcinoma and its relation to chronic kidney disease in genotypically at-risk African ancestry individuals. Abstract Although the relative risk of renal cell carcinoma associated with chronic kidney injury is particularly high among sub-Saharan African ancestry populations, it is unclear yet whether the APOL1 gene risk variants (RV) for kidney disease additionally elevate this risk. APOL1 G1 and G2 RV contribute to increased risk for kidney disease in black populations, although the disease mechanism has still not been fully deciphered. While high expression levels of all three APOL1 allelic variants, G0 (the wild type allele), G1, and G2 are injurious to normal human cells, renal carcinoma cells (RCC) naturally tolerate inherent high expression levels of APOL1. We utilized CRISPR/Cas9 gene editing to generate isogenic RCC clones expressing APOL1 G1 or G2 risk variants on a similar genetic background, thus enabling a reliable comparison between the phenotypes elicited in RCC by each of the APOL1 variants. Here, we demonstrate that knocking in the G1 or G2 APOL1 alleles, or complete elimination of APOL1 expression, has major effects on proliferation capacity, mitochondrial morphology, cell metabolism, autophagy levels, and the tumorigenic potential of RCC cells. The most striking effect of the APOL1 RV effect was demonstrated in vivo by the complete abolishment of tumor growth in immunodeficient mice. Our findings suggest that, in contrast to the WT APOL1 variant, APOL1 RV are toxic for RCC cells and may act to suppress cancer cell growth. We conclude that the inherent expression of non-risk APOL1 G0 is required for RCC tumorigenicity. RCC cancer cells can hardly tolerate increased APOL1 risk variants expression levels as opposed to APOL1 G0.
Collapse
|
38
|
Ruhl AP, Jeffries N, Yang Y, Naik RP, Patki A, Pecker LH, Mott BT, Zakai NA, Winkler CA, Kopp JB, Lange LA, Irvin MR, Gutierrez OM, Cushman M, Ackerman HC. Alpha Globin Gene Copy Number Is Associated with Prevalent Chronic Kidney Disease and Incident End-Stage Kidney Disease among Black Americans. J Am Soc Nephrol 2022; 33:213-224. [PMID: 34706968 PMCID: PMC8763181 DOI: 10.1681/asn.2021050653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/05/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND α-Globin is expressed in endothelial cells of resistance arteries, where it limits endothelial nitric oxide signaling and enhances α-adrenergic-mediated vasoconstriction. α-Globin gene (HBA) copy number is variable in people of African descent and other populations worldwide. Given the protective effect of nitric oxide in the kidney, we hypothesized that HBA copy number would be associated with kidney disease risk. METHODS Community-dwelling Black Americans aged ≥45 years old were enrolled in a national longitudinal cohort from 2003 through 2007. HBA copy number was measured using droplet digital PCR. The prevalence ratio (PR) of CKD and the relative risk (RR) of incident reduced eGFR were calculated using modified Poisson multivariable regression. The hazard ratio (HR) of incident ESKD was calculated using Cox proportional hazards multivariable regression. RESULTS Among 9908 participants, HBA copy number varied from 2 to 6. In analyses adjusted for demographic, clinical, and genetic risk factors, a one-copy increase in HBA was associated with 14% greater prevalence of CKD (PR, 1.14; 95% CI, 1.07 to 1.21; P<0.0001). While HBA copy number was not associated with incident reduced eGFR (RR, 1.06; 95% CI, 0.94 to 1.19; P=0.38), the hazard of incident ESKD was 32% higher for each additional copy of HBA (HR, 1.32; 95% CI, 1.09 to 1.61; P=0.005). CONCLUSIONS Increasing HBA copy number was associated with a greater prevalence of CKD and incidence of ESKD in a national longitudinal cohort of Black Americans.
Collapse
Affiliation(s)
- A. Parker Ruhl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland,Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Yu Yang
- Division of Blood Diseases and Resources, National Heart, Lung, and Blood Institute, Rockville, Maryland
| | - Rakhi P. Naik
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amit Patki
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lydia H. Pecker
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bryan T. Mott
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Neil A. Zakai
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont,Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| | - Cheryl A. Winkler
- Basic Research Program, National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jeffrey B. Kopp
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado, Denver, Colorado
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama
| | - Orlando M. Gutierrez
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama,Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Mary Cushman
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont,Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| | - Hans C. Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
39
|
Abstract
The parasitic trypanosomatids cause lethal and debilitating diseases, the leishmaniases, Chagas disease, and the African trypanosomiases, with major impacts on human and animal health. Sustained research has borne fruit by assisting efforts to reduce the burden of disease and by improving our understanding of fundamental molecular and cell biology. But where has the research primarily been conducted, and which research areas have received the most attention? These questions are addressed below using publication and citation data from the past few decades.
Collapse
Affiliation(s)
- David Horn
- The Wellcome Trust Centre for Anti-Infectives Research, Division of Biological Chemistry & Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
40
|
Müller D, Schmitz J, Fischer K, Granado D, Groh AC, Krausel V, Lüttgenau SM, Amelung TM, Pavenstädt H, Weide T. Evolution of Renal-Disease Factor APOL1 Results in Cis and Trans Orientations at the Endoplasmic Reticulum That Both Show Cytotoxic Effects. Mol Biol Evol 2021; 38:4962-4976. [PMID: 34323996 PMCID: PMC8557400 DOI: 10.1093/molbev/msab220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The recent and exclusively in humans and a few other higher primates expressed APOL1 (apolipoprotein L1) gene is linked to African human trypanosomiasis (also known as African sleeping sickness) as well as to different forms of kidney diseases. Whereas APOL1's role as a trypanolytic factor is well established, pathobiological mechanisms explaining its cytotoxicity in renal cells remain unclear. In this study, we compared the APOL family members using a combination of evolutionary studies and cell biological experiments to detect unique features causal for APOL1 nephrotoxic effects. We investigated available primate and mouse genome and transcriptome data to apply comparative phylogenetic and maximum likelihood selection analyses. We suggest that the APOL gene family evolved early in vertebrates and initial splitting occurred in ancestral mammals. Diversification and differentiation of functional domains continued in primates, including developing the two members APOL1 and APOL2. Their close relationship could be diagnosed by sequence similarity and a shared ancestral insertion of an AluY transposable element. Live-cell imaging analyses showed that both expressed proteins show a strong preference to localize at the endoplasmic reticulum (ER). However, glycosylation and secretion assays revealed that-unlike APOL2-APOL1 membrane insertion or association occurs in different orientations at the ER, with the disease-associated mutants facing either the luminal (cis) or cytoplasmic (trans) side of the ER. The various pools of APOL1 at the ER offer a novel perspective in explaining the broad spectrum of its observed toxic effects.
Collapse
Affiliation(s)
- Daria Müller
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Jürgen Schmitz
- Institute of Experimental Pathology, ZMBE, University of Münster, Münster, Germany
| | - Katharina Fischer
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Daniel Granado
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Ann-Christin Groh
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Vanessa Krausel
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Simona Mareike Lüttgenau
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Till Maximilian Amelung
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Hermann Pavenstädt
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Thomas Weide
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| |
Collapse
|
41
|
Masimango MI, Jadoul M, Binns-Roemer EA, David VA, Sumaili EK, Winkler CA, Limou S. APOL1 Renal Risk Variants and Sickle Cell Trait Associations With Reduced Kidney Function in a Large Congolese Population-Based Study. Kidney Int Rep 2021; 7:474-482. [PMID: 35257060 PMCID: PMC8897685 DOI: 10.1016/j.ekir.2021.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
Affiliation(s)
- Mannix Imani Masimango
- Department of Internal Medicine, Hôpital Provincial Général de Référence de Bukavu, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Michel Jadoul
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Elizabeth A. Binns-Roemer
- Basic Science Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Victor A. David
- Basic Science Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Ernest Kiswaya Sumaili
- Department of Nephrology, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Cheryl A. Winkler
- Basic Science Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Sophie Limou
- Basic Science Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland, USA
- Institute for Transplantation in Urology-Nephrology, Centre de Recherche en Transplantation et Immunologie, UMR1064, Institut National de la Santé et de la Recherche Médicale, Université de Nantes, Nantes, France
- Ecole Centrale de Nantes, Computer sciences and Mathematics in Biology Department, Nantes, France
- Correspondence: Sophie Limou, Centre de Recherche en Transplantation et Immunologie, Institut National de la Santé et de la Recherche Médicale, UMR1064 - Team 5, 30 bd Jean Monnet, 44093 Nantes Cedex 1, France.
| |
Collapse
|
42
|
Pant J, Samanovic M, Nelson MT, Keceli MK, Verdi J, Beverley SM, Raper J. Interplay of Trypanosome Lytic Factor and innate immune cells in the resolution of cutaneous Leishmania infection. PLoS Pathog 2021; 17:e1008768. [PMID: 34559857 PMCID: PMC8494325 DOI: 10.1371/journal.ppat.1008768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/06/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
Trypanosome Lytic Factor (TLF) is a primate-specific high-density lipoprotein (HDL) complex that, through the cation channel-forming protein apolipoprotein L-1 (APOL1), provides innate immunity to select kinetoplastid parasites. The immunoprotective effects of TLF have been extensively investigated in the context of its interaction with the extracellular protozoan Trypanosoma brucei brucei, to which it confers sterile immunity. We previously showed that TLF could act against an intracellular pathogen Leishmania, and here we dissected the role of TLF and its synergy with host-immune cells. Leishmania major is transmitted by Phlebotomine sand flies, which deposit the parasite intradermally into mammalian hosts, where neutrophils are the predominant phagocytes recruited to the site of infection. Once in the host, the parasites are phagocytosed and shed their surface glycoconjugates during differentiation to the mammalian-resident amastigote stage. Our data show that mice producing TLF have reduced parasite burdens when infected intradermally with metacyclic promastigotes of L. major, the infective, fly-transmitted stage. This TLF-mediated reduction in parasite burden was lost in neutrophil-depleted mice, suggesting that early recruitment of neutrophils is required for TLF-mediated killing of L. major. In vitro we find that only metacyclic promastigotes co-incubated with TLF in an acidic milieu were lysed. However, amastigotes were not killed by TLF at any pH. These findings correlated with binding experiments, revealing that labeled TLF binds specifically to the surface of metacyclic promastigotes, but not to amastigotes. Metacyclic promastigotes of L. major deficient in the synthesis of surface glycoconjugates LPG and/or PPG (lpg1- and lpg5A-/lpg5B- respectively) whose absence mimics the amastigote surface, were resistant to TLF-mediated lysis. We propose that TLF binds to the outer surface glycoconjugates of metacyclic promastigotes, whereupon it kills the parasite in the acidic phagosome of phagocytes. We hypothesize that resistance to TLF requires shedding of the surface glycoconjugates, which occurs several hours after phagocytosis by immune cells, creating a relatively short-lived but effective window for TLF to act against Leishmania. Leishmaniasis, the disease caused by parasites of the genus Leishmania, can be divided into cutaneous, muco-cutaneous and visceral leishmaniasis depending on the parasite species and the clinical outcome of the disease. Of the three, cutaneous leishmaniasis is the most common form, which is usually characterized by a localized lesion due to the infection of immune cells, primarily dermal and lymph node-resident macrophages. The time between infection and lesion appearance ranges from weeks to years, while some individuals never develop lesions. The length of this subclinical stage of leishmaniasis depends on a variety of factors: parasite virulence, infectious dose, and the host immune response. Therefore, it remains crucial to develop our understanding of each component of the host-parasite interface and assess the role that each component plays in the clinical outcome. Here, we analyze the interaction between L. major, a cutaneous strain, and the host innate immune factor Trypanosome Lytic Factor (TLF), a sub-class of circulating High-Density Lipoprotein (HDL). TLF provides sterile immunity to most extracellular African Trypanosomes by osmotically lysing the parasites. Lysis is driven by the primate specific protein apolipoprotein L-1 (APOL1), a cation channel-forming protein that is activated by a series of pH-dependent conformational changes. APOL1 inserts into cellular membranes at acidic pH and forms a closed ion channel that subsequently opens when re-exposed to neutral pH, resulting in ion flux. Using transgenic mice producing primate TLF, we show that both human and baboon TLFs ameliorate cutaneous Leishmania major infection and that this reduction in parasite burden correlates with: 1. infectious dose of metacyclic promastigotes 2. the concentration of circulating TLF in plasma and 3. early recruitment of neutrophils at the site of infection. Our results show that the acidification step is essential for TLF-mediated lysis of axenic metacyclic promastigotes of Leishmania in vitro. The susceptibility of metacyclic promastigotes to TLF-mediated lysis is governed by the surface glycoconjugates of Leishmania. We find that surface glycoconjugate-deficient Leishmania are resistant to TLF-mediated killing. Based on these data, we conclude that the shedding of surface glycoconjugates while transitioning from metacyclic promastigotes to amastigotes, results in parasite resistance to TLF-mediated lysis. Whether TLF is effective at killing metacyclic promastigotes of other experimentally tractable Leishmania sp., such as L. infantum and L. donovani, which have different surface glycoconjugate structures is yet to be tested. Our data raise the possibility that TLF may have lytic activity against a broader range of pathogens such as bacteria, viruses, fungi and parasites with surface glycoconjugates that transit through intracellular acidic compartments.
Collapse
Affiliation(s)
- Jyoti Pant
- Department of Biology, Hunter College, City University of New York, New York, New York, United States of America
- Molecular, Cellular and Developmental biology, The Graduate Center, City University of New York, New York, New York, United States of America
- * E-mail: (JP); (JR)
| | - Marie Samanovic
- Medical Parasitology, New York University Langone Medical Center, New York, New York, United States of America
| | - Maria T. Nelson
- Department of Biology, Hunter College, City University of New York, New York, New York, United States of America
| | - Mert K. Keceli
- Department of Biology, Hunter College, City University of New York, New York, New York, United States of America
| | - Joseph Verdi
- Department of Biology, Hunter College, City University of New York, New York, New York, United States of America
- Molecular, Cellular and Developmental biology, The Graduate Center, City University of New York, New York, New York, United States of America
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jayne Raper
- Department of Biology, Hunter College, City University of New York, New York, New York, United States of America
- Molecular, Cellular and Developmental biology, The Graduate Center, City University of New York, New York, New York, United States of America
- * E-mail: (JP); (JR)
| |
Collapse
|
43
|
Machado H, Bizarra-Rebelo T, Costa-Sequeira M, Trindade S, Carvalho T, Rijo-Ferreira F, Rentroia-Pacheco B, Serre K, Figueiredo LM. Trypanosoma brucei triggers a broad immune response in the adipose tissue. PLoS Pathog 2021; 17:e1009933. [PMID: 34525131 PMCID: PMC8476018 DOI: 10.1371/journal.ppat.1009933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/27/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is one of the major reservoirs of Trypanosoma brucei parasites, the causative agent of sleeping sickness, a fatal disease in humans. In mice, the gonadal adipose tissue (AT) typically harbors 2–5 million parasites, while most solid organs show 10 to 100-fold fewer parasites. In this study, we tested whether the AT environment responds immunologically to the presence of the parasite. Transcriptome analysis of T. brucei infected adipose tissue revealed that most upregulated host genes are involved in inflammation and immune cell functions. Histochemistry and flow cytometry confirmed an increasingly higher number of infiltrated macrophages, neutrophils and CD4+ and CD8+ T lymphocytes upon infection. A large proportion of these lymphocytes effectively produce the type 1 effector cytokines, IFN-γ and TNF-α. Additionally, the adipose tissue showed accumulation of antigen-specific IgM and IgG antibodies as infection progressed. Mice lacking T and/or B cells (Rag2-/-, Jht-/-), or the signature cytokine (Ifng-/-) displayed a higher parasite load both in circulation and in the AT, demonstrating the key role of the adaptive immune system in both compartments. Interestingly, infections of C3-/- mice showed that while complement system is dispensable to control parasite load in the blood, it is necessary in the AT and other solid tissues. We conclude that T. brucei infection triggers a broad and robust immune response in the AT, which requires the complement system to locally reduce parasite burden. African trypanosomiasis is a neglected disease with significant socio-economic burden in sub-Saharan Africa. The protozoan parasite Trypanosoma brucei, a causative agent of African trypanosomiasis, can be found in the blood and extra-vascular spaces of the infected host. For an unknown reason, T. brucei accumulates in adipose tissue (AT) in very high numbers. Here we used a multidisciplinary approach to assess whether an immune response was mounted in AT during a T. brucei infection. We found that as infection progresses, a broad variety of immune cells and antibodies accumulate in the AT. We also found that this broad immune response is partially able to control parasite numbers in the AT. Our study provides evidence that T. brucei parasites present in the AT are subjected to immune surveillance. The reason why T. brucei accumulates to such a high extent in AT remains to be elucidated.
Collapse
Affiliation(s)
- Henrique Machado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago Bizarra-Rebelo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana Costa-Sequeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sandra Trindade
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Barbara Rentroia-Pacheco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- * E-mail: (KS); (LMF)
| | - Luisa M. Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- * E-mail: (KS); (LMF)
| |
Collapse
|
44
|
Silva Pereira S, Jackson AP, Figueiredo LM. Evolution of the variant surface glycoprotein family in African trypanosomes. Trends Parasitol 2021; 38:23-36. [PMID: 34376326 DOI: 10.1016/j.pt.2021.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023]
Abstract
An intriguing and remarkable feature of African trypanosomes is their antigenic variation system, mediated by the variant surface glycoprotein (VSG) family and fundamental to both immune evasion and disease epidemiology within host populations. Recent studies have revealed that the VSG repertoire has a complex evolutionary history. Sequence diversity, genomic organization, and expression patterns are species-specific, which may explain other variations in parasite virulence and disease pathology. Evidence also shows that we may be underestimating the extent to what VSGs are repurposed beyond their roles as variant antigens, establishing a need to examine VSG functionality more deeply. Here, we review sequence variation within the VSG gene family, and highlight the many opportunities to explore their likely diverse contributions to parasite survival.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Andrew P Jackson
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 5RF, UK
| | - Luísa M Figueiredo
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
45
|
Schaub C, Lee P, Racho-Jansen A, Giovinazzo J, Terra N, Raper J, Thomson R. Coiled-coil binding of the leucine zipper domains of APOL1 is necessary for the open cation channel conformation. J Biol Chem 2021; 297:101009. [PMID: 34331942 PMCID: PMC8446801 DOI: 10.1016/j.jbc.2021.101009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/12/2021] [Accepted: 07/23/2021] [Indexed: 11/17/2022] Open
Abstract
Apolipoprotein L-I (APOL1) is a channel-forming effector of innate immunity. The common human APOL1 variant G0 provides protection against infection with certain Trypanosoma and Leishmania parasite species, but it cannot protect against the trypanosomes responsible for human African trypanosomiasis. Human APOL1 variants G1 and G2 protect against human-infective trypanosomes but also confer a higher risk of developing chronic kidney disease. Trypanosome-killing activity is dependent on the ability of APOL1 to insert into membranes at acidic pH and form pH-gated cation channels. We previously mapped the channel’s pore-lining region to the C-terminal domain (residues 332–398) and identified a membrane-insertion domain (MID, residues 177–228) that facilitates acidic pH-dependent membrane insertion. In this article, we further investigate structural determinants of cation channel formation by APOL1. Using a combination of site-directed mutagenesis and targeted chemical modification, our data indicate that the C-terminal heptad-repeat sequence (residues 368–395) is a bona fide leucine zipper domain (ZIP) that is required for cation channel formation as well as lysis of trypanosomes and mammalian cells. Using protein-wide cysteine-scanning mutagenesis, coupled with the substituted cysteine accessibility method, we determined that, in the open channel state, both the N-terminal domain and the C-terminal ZIP domain are exposed on the intralumenal/extracellular side of the membrane and provide evidence that each APOL1 monomer contributes four transmembrane domains to the open cation channel conformation. Based on these data, we propose an oligomeric topology model in which the open APOL1 cation channel is assembled from the coiled-coil association of C-terminal ZIP domains.
Collapse
Affiliation(s)
- Charles Schaub
- Department of Biological sciences, Hunter College, City University of New York, USA; The Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York; Vanderbilt University, Nashville, Tennessee, USA
| | - Penny Lee
- Department of Biological sciences, Hunter College, City University of New York, USA; John Jay College, City University of New York, USA
| | - Alisha Racho-Jansen
- Department of Biological sciences, Hunter College, City University of New York, USA
| | - Joe Giovinazzo
- Department of Biological sciences, Hunter College, City University of New York, USA; University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nada Terra
- Department of Biological sciences, Hunter College, City University of New York, USA; Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jayne Raper
- Department of Biological sciences, Hunter College, City University of New York, USA; The Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York.
| | - Russell Thomson
- Department of Biological sciences, Hunter College, City University of New York, USA.
| |
Collapse
|
46
|
Ekulu PM, Adebayo OC, Decuypere JP, Bellucci L, Elmonem MA, Nkoy AB, Mekahli D, Bussolati B, van den Heuvel LP, Arcolino FO, Levtchenko EN. Novel Human Podocyte Cell Model Carrying G2/G2 APOL1 High-Risk Genotype. Cells 2021; 10:cells10081914. [PMID: 34440683 PMCID: PMC8391400 DOI: 10.3390/cells10081914] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 02/01/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk genotypes (HRG), G1 and G2, increase the risk of various non-diabetic kidney diseases in the African population. To date, the precise mechanisms by which APOL1 risk variants induce injury on podocytes and other kidney cells remain unclear. Trying to unravel these mechanisms, most studies have used animal or cell models created by gene editing. We developed and characterised conditionally immortalised human podocyte cell lines derived from urine of a donor carrying APOL1 HRG G2/G2. Following induction of APOL1 expression by polyinosinic-polycytidylic acid (poly(I:C)), we assessed functional features of APOL1-induced podocyte dysfunction. As control, APOL1 wild type (G0/G0) podocyte cell line previously generated from a Caucasian donor was used. Upon exposure to poly(I:C), G2/G2 and G0/G0 podocytes upregulated APOL1 expression resulting in podocytes detachment, decreased cells viability and increased apoptosis rate in a genotype-independent manner. Nevertheless, G2/G2 podocyte cell lines exhibited altered features, including upregulation of CD2AP, alteration of cytoskeleton, reduction of autophagic flux and increased permeability in an in vitro model under continuous perfusion. The human APOL1 G2/G2 podocyte cell model is a useful tool for unravelling the mechanisms of APOL1-induced podocyte injury and the cellular functions of APOL1.
Collapse
Affiliation(s)
- Pepe M. Ekulu
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Department of Paediatrics, Division of Nephrology, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Oyindamola C. Adebayo
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Jean-Paul Decuypere
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
| | - Linda Bellucci
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10124 Turin, Italy; (L.B.); (B.B.)
| | - Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 11628, Egypt;
| | - Agathe B. Nkoy
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Department of Paediatrics, Division of Nephrology, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Djalila Mekahli
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Department of Paediatrics, Division of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10124 Turin, Italy; (L.B.); (B.B.)
| | - Lambertus P. van den Heuvel
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Department of Paediatric Nephrology, Radboud University Medical Centre, 6500 Nijmegen, The Netherlands
| | - Fanny O. Arcolino
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Correspondence: ; Tel.: +32-16372647
| | - Elena N. Levtchenko
- Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (P.M.E.); (O.C.A.); (J.-P.D.); (A.B.N.); (D.M.); (L.P.v.d.H.); (E.N.L.)
- Department of Paediatrics, Division of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
47
|
Ultsch M, Holliday MJ, Gerhardy S, Moran P, Scales SJ, Gupta N, Oltrabella F, Chiu C, Fairbrother W, Eigenbrot C, Kirchhofer D. Structures of the ApoL1 and ApoL2 N-terminal domains reveal a non-classical four-helix bundle motif. Commun Biol 2021; 4:916. [PMID: 34316015 PMCID: PMC8316464 DOI: 10.1038/s42003-021-02387-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein L1 (ApoL1) is a circulating innate immunity protein protecting against trypanosome infection. However, two ApoL1 coding variants are associated with a highly increased risk of chronic kidney disease. Here we present X-ray and NMR structures of the N-terminal domain (NTD) of ApoL1 and of its closest relative ApoL2. In both proteins, four of the five NTD helices form a four-helix core structure which is different from the classical four-helix bundle and from the pore-forming domain of colicin A. The reactivity with a conformation-specific antibody and structural models predict that this four-helix motif is also present in the NTDs of ApoL3 and ApoL4, suggesting related functions within the small ApoL family. The long helix 5 of ApoL1 is conformationally flexible and contains the BH3-like region. This BH3-like α-helix resembles true BH3 domains only in sequence and structure but not in function, since it does not bind to the pro-survival members of the Bcl-2 family, suggesting a Bcl-2-independent role in cytotoxicity. These findings should expedite a more comprehensive structural and functional understanding of the ApoL immune protein family.
Collapse
Affiliation(s)
- Mark Ultsch
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Michael J Holliday
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Stefan Gerhardy
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Paul Moran
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Suzie J Scales
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Nidhi Gupta
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | | | - Cecilia Chiu
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Wayne Fairbrother
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Charles Eigenbrot
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
48
|
Gaudet RG, Zhu S, Halder A, Kim BH, Bradfield CJ, Huang S, Xu D, Mamiñska A, Nguyen TN, Lazarou M, Karatekin E, Gupta K, MacMicking JD. A human apolipoprotein L with detergent-like activity kills intracellular pathogens. Science 2021; 373:eabf8113. [PMID: 34437126 PMCID: PMC8422858 DOI: 10.1126/science.abf8113] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.
Collapse
Affiliation(s)
- Ryan G Gaudet
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shiwei Zhu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Anushka Halder
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Bae-Hoon Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Clinton J Bradfield
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shuai Huang
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Dijin Xu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Agnieszka Mamiñska
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thanh Ngoc Nguyen
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Erdem Karatekin
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Saints-Pères Paris Institute for the Neurosciences, Centre National de la Recherche Scientifique (CNRS), Université de Paris, F-75006 Paris, France
| | - Kallol Gupta
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
49
|
Dean S. Basic Biology of Trypanosoma brucei with Reference to the Development of Chemotherapies. Curr Pharm Des 2021; 27:1650-1670. [PMID: 33463458 DOI: 10.2174/1381612827666210119105008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 11/22/2022]
Abstract
Trypanosoma brucei are protozoan parasites that cause the lethal human disease African sleeping sickness and the economically devastating disease of cattle, Nagana. African sleeping sickness, also known as Human African Trypanosomiasis (HAT), threatens 65 million people and animal trypanosomiasis makes large areas of farmland unusable. There is no vaccine and licensed therapies against the most severe, late-stage disease are toxic, impractical and ineffective. Trypanosomes are transmitted by tsetse flies, and HAT is therefore predominantly confined to the tsetse fly belt in sub-Saharan Africa. They are exclusively extracellular and they differentiate between at least seven developmental forms that are highly adapted to host and vector niches. In the mammalian (human) host they inhabit the blood, cerebrospinal fluid (late-stage disease), skin, and adipose fat. In the tsetse fly vector they travel from the tsetse midgut to the salivary glands via the ectoperitrophic space and proventriculus. Trypanosomes are evolutionarily divergent compared with most branches of eukaryotic life. Perhaps most famous for their extraordinary mechanisms of monoallelic gene expression and antigenic variation, they have also been investigated because much of their biology is either highly unconventional or extreme. Moreover, in addition to their importance as pathogens, many researchers have been attracted to the field because trypanosomes have some of the most advanced molecular genetic tools and database resources of any model system. The following will cover just some aspects of trypanosome biology and how its divergent biochemistry has been leveraged to develop drugs to treat African sleeping sickness. This is by no means intended to be a comprehensive survey of trypanosome features. Rather, I hope to present trypanosomes as one of the most fascinating and tractable systems to do discovery biology.
Collapse
Affiliation(s)
- Samuel Dean
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| |
Collapse
|
50
|
Lack of APOL1 in proximal tubules of normal human kidneys and proteinuric APOL1 transgenic mouse kidneys. PLoS One 2021; 16:e0253197. [PMID: 34138902 PMCID: PMC8211208 DOI: 10.1371/journal.pone.0253197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/30/2021] [Indexed: 11/19/2022] Open
Abstract
The mechanism of pathogenesis associated with APOL1 polymorphisms and risk for non-diabetic chronic kidney disease (CKD) is not fully understood. Prior studies have minimized a causal role for the circulating APOL1 protein, thus efforts to understand kidney pathogenesis have focused on APOL1 expressed in renal cells. Of the kidney cells reported to express APOL1, the proximal tubule expression patterns are inconsistent in published reports, and whether APOL1 is synthesized by the proximal tubule or possibly APOL1 protein in the blood is filtered and reabsorbed by the proximal tubule remains unclear. Using both protein and mRNA in situ methods, the kidney expression pattern of APOL1 was examined in normal human and APOL1 bacterial artificial chromosome transgenic mice with and without proteinuria. APOL1 protein and mRNA was detected in podocytes and endothelial cells, but not in tubular epithelia. In the setting of proteinuria, plasma APOL1 protein did not appear to be filtered or reabsorbed by the proximal tubule. A side-by-side examination of commercial antibodies used in prior studies suggest the original reports of APOL1 in proximal tubules likely reflects antibody non-specificity. As such, APOL1 expression in podocytes and endothelia should remain the focus for mechanistic studies in the APOL1-mediated kidney diseases.
Collapse
|