Modulation of Cystatin C in Human Macrophages Improves Anti-Mycobacterial Immune Responses to Mycobacterium tuberculosis Infection and Coinfection With HIV

Tuberculosis owes its resurgence as a major global health threat mostly to the emergence of drug resistance and coinfection with HIV. The synergy between HIV and Mycobacterium tuberculosis (Mtb) modifies the host immune environment to enhance both viral and bacterial replication and spread. In the lung immune context, both pathogens infect macrophages, establishing favorable intracellular niches. Both manipulate the endocytic pathway in order to avoid destruction. Relevant players of the endocytic pathway to control pathogens include endolysosomal proteases, cathepsins, and their natural inhibitors, cystatins.
Here, a mapping of the human macrophage transcriptome for type I and II cystatins during Mtb, HIV, or Mtb-HIV infection displayed different profiles of gene expression, revealing cystatin C as a potential target to control mycobacterial infection as well as HIV coinfection. We found that cystatin C silencing in macrophages significantly improves the intracellular killing of Mtb, which was concomitant with an increased general proteolytic activity of cathepsins. In addition, downmodulation of cystatin C led to an improved expression of the human leukocyte antigen (HLA) class II in macrophages and an increased CD4+ T-lymphocyte proliferation along with enhanced IFN-γ secretion. Overall, our results suggest that the targeting of cystatin C in human macrophages represents a promising approach to improve the control of mycobacterial infections including multidrug-resistant (MDR) TB.

piggyBac Transposition and the Expression of Human Cystatin C in Transgenic Chickens

A bioreactor can be used for mass production of therapeutic proteins and other bioactive substances. Although various methods have been developed using microorganisms and animal cells, advanced strategies are needed for the efficient production of biofunctional proteins. In microorganisms, post-translational glycosylation and modification are not performed properly, while animal cell systems require more time and expense. To overcome these problems, new methods using products from transgenic animals have been considered, such as genetically modified cow’s milk and hen’s eggs. In this study, based on a non-viral piggyBac transposition system, we generated transgenic bioreactor chickens that produced human cystatin C (hCST3).
There were no differences in the phenotype or histochemical structure of the wild-type and hCST3-expressing transgenic chickens. Subsequently, we analyzed the hCST3 expression in transgenic chickens, mainly in muscle and egg white, which could be major deposition warehouses for hCST3 protein. In both muscle and egg white, we detected high hCST3 expression by ELISA and Western blotting. hCST3 proteins were efficiently purified from muscle and egg white of transgenic chickens using a His-tag purification system. These data show that transgenic chickens can be efficiently used as a bioreactor for the mass production of bioactive materials.

DMPC Phospholipid Bilayer as a Potential Interface for Human Cystatin C Oligomerization: Analysis of Protein-Liposome Interactions Using NMR Spectroscopy

Studies revolving around mechanisms responsible for the development of amyloid-based diseases lay the foundations for the recognition of molecular targets of future to-be-developed treatments. However, the vast number of peptides and proteins known to be responsible for fibril formation, combined with their complexity and complexity of their interactions with various cellular components, renders this task extremely difficult and time-consuming. One of these proteins, human cystatin C (hCC), is a well-known and studied cysteine-protease inhibitor. While being a monomer in physiological conditions, under the necessary stimulus-usually a mutation, it tends to form fibrils, which later participate in the disease development.
This process can potentially be regulated (in several ways) by many cellular components and it is being hypothesized that the cell membrane might play a key role in the oligomerization pathway. Studies involving cell membranes pose several difficulties; therefore, an alternative in the form of membrane mimetics is a very attractive solution. Here, we would like to present the first study on hCC oligomerization under the influence of phospholipid liposomes, acting as a membrane mimetic. The protein-mimetic interactions are studied utilizing circular dichroism, nuclear magnetic resonance, and size exclusion chromatography.

Microfluidic electrochemical immunosensor for the determination of cystatin C in human serum

  • The fabrication of a nanointerfaced electrochemical immunosensor is described for the rapid determination of cystatin C, a biomarker that is elevated in diabetic retinopathy. A dispersion of graphene oxide-chitosan (GO-Chit) nanocomposite was used to modify the carbon working electrode, allowing for a high conjugation of anti-cystatin C antibody. This modified sensor was characterized both morphologically and electrochemically, and the sensor performance was evaluated towards selective quantification of cystatin C in simulated as well as serum samples using cyclic voltammetry and differential pulse voltammetry.
  • The sensor was able to detect cystatin C in the concentration range1 – 10 mg/L with a detection limit of 0.0078 mg/L. The preparation time of the sensor was 420 s, which was faster than that of conventional ELISA and other electrochemical sensors reported in literature. The clinical applicability of the proposed electrochemical biosensor was demonstrated through quantification of cystatin C in human serum samples and identification of diabetic retinopathy. A cutoff value of 1.2 mg/L of cystatin C was used beyond which the samples were classified as positive for diabetic retinopathy.
  • Two different working electrodes, namely a glassy carbon electrode (GCE) and paper electrodes, were used in the study. The working potential was set to 0.25 V vs. Ag/AgCl for experiments with the GCE and 0.15 V for the paper electrodes. The prediction was validated by clinical diagnosis wherein the prediction accuracy of the sensor exceeded 85%. The sensor platform was translated onto a paper substrate and characterized for achieving an optimum sensing performance. This work is the first attempt to employ an electrochemical cystatin C sensor for the diagnosis of diabetic retinopathy from serum samples. Graphical abstract.

OPPA01325-20UG - Cystatin C Human - Human Cystatin C Protein

OPPA01325-20UG Aviva Systems Biology 20ug 85 EUR

Human Cystatin C

7-04936 CHI Scientific $20µg Ask for price

Human Cystatin C

7-04937 CHI Scientific 100µg Ask for price

Human Cystatin C

7-04938 CHI Scientific 1mg Ask for price

Cystatin-C Human

22060945-1 Glycomatrix 20 µg 137.27 EUR

Cystatin-C Human

rAP-3125 Angio Proteomie Inquiry Ask for price

Cystatin C, Human urine

P1447-100 Biovision each 405.6 EUR

Cystatin C (Human) ELISA Kit

EKA52036-5x96T Biomatik Corporation 5x96T Ask for price

Cystatin C (Human) ELISA Kit

EKA52036-96T Biomatik Corporation 96T 446.9 EUR

Human Cystatin C ELISA

1041 Polysciences Europe GmbH - 567 EUR

Human Cystatin C ELISA

GWB-374CYS GenWay Biotech 96 well Ask for price

Human Cystatin C Protein

abx061016-1mg Abbexa 1 mg 886.8 EUR

Human Cystatin C Protein

20-abx060110 Abbexa
  • Ask for price
  • Ask for price
  • 100 ug
  • 1 mg

Human Cystatin C Protein

abx060233-1mg Abbexa 1 mg 927.6 EUR

Human Cystatin C Protein

abx060714-1mg Abbexa 1 mg 844.8 EUR

Human Cystatin-C Protein

abx060716-1mg Abbexa 1 mg 627.6 EUR

Human Cystatin C Antibody

33137-05111 AssayPro 150 ug 313.2 EUR

human Cystatin-C protein

CSB-NP061941h Cusabio 10mg Ask for price

Cystatin C Antibody / Cystatin 3

RQ4449 NSJ Bioreagents 100ug 356.15 EUR

Cystatin C Antibody / Cystatin 3

RQ4450 NSJ Bioreagents 100ug 356.15 EUR

Cystatin C Antibody / Cystatin 3

RQ4858 NSJ Bioreagents 100ul 356.15 EUR

Cystatin C Antibody / Cystatin 3

RQ4885 NSJ Bioreagents 100 ug 356.15 EUR

Structural Characterization of Covalently Stabilized Human Cystatin C Oligomers

Human cystatin C (HCC), a cysteine-protease inhibitor, exists as a folded monomer under physiological conditions but has the ability to self-assemble via domain swapping into multimeric states, including oligomers with a doughnut-like structure. The structure of the monomeric HCC has been solved by X-ray crystallography, and a covalently linked version of HCC (stab-1 HCC) is able to form stable oligomeric species containing 10-12 monomeric subunits. We have performed molecular modeling, and in conjunction with experimental parameters obtained from atomic force microscopy (AFM), transmission electron microscopy (TEM) and small-angle X-ray scattering (SAXS) measurements, we observe that the structures are essentially flat, with a height of about 2 nm, and the distance between the outer edge of the ring and the edge of the central cavity is ~5.1 nm.
These dimensions correspond to the height and diameter of one stab-1 HCC subunit and we present a dodecamer model for stabilized cystatin C oligomers using molecular dynamics simulations and experimentally measured parameters. Given that oligomeric species in protein aggregation reactions are often transient and very highly heterogeneous, the structural information presented here on these isolated stab-1 HCC oligomers may be useful to further explore the physiological relevance of different structural species of cystatin C in relation to protein misfolding disease.

Leave a Reply

Your email address will not be published.