Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ziqing Jiang is active.

Publication


Featured researches published by Ziqing Jiang.


Biopolymers | 2008

Effects of net charge and the number of positively charged residues on the biological activity of amphipathic α-helical cationic antimicrobial peptides

Ziqing Jiang; Adriana I. Vasil; John D. Hale; Robert E. W. Hancock; Michael L. Vasil; Robert S. Hodges

In our previous study, we utilized a 26‐residue amphipathic α‐helical antimicrobial peptide L‐V13K (Chen et al., Antimicrob Agents Chemother 2007, 51, 1398–1406) as the framework to study the effects of peptide hydrophobicity on the mechanism of its antimicrobial action. In this study, we explored the effects of net charge and the number of positively charged residues on the hydrophilic/polar face of L‐V13K on its biological activity (antimicrobial and hemolytic) and biophysical properties (hydrophobicity, amphipathicity, helicity, and peptide self‐association). The net charge of V13K analogs at pH 7 varied between −5 and +10 and the number of positively charged residues varied from 1 to 10. The minimal inhibitory concentrations (MIC) against six strains of Pseudomonas aeruginosa as well as other gram‐negative and gram‐positive bacteria were determined along with the maximal peptide concentration that produces no hemolysis of human red blood cells (MHC). Our results show that the number of positively charged residues on the polar face and net charge are both important for both antimicrobial activity and hemolytic activity. The most dramatic observation is the sharp transition of hemolytic activity on increasing one positive charge on the polar face of V13K i.e., the change from +8 to +9 resulted in greater than 32‐fold increase in hemolytic activity (250 μg/ml to <7.8 μg/ml, respectively).


Chemical Biology & Drug Design | 2011

Rational design of α-helical antimicrobial peptides to target Gram-negative pathogens, Acinetobacter baumannii and Pseudomonas aeruginosa: utilization of charge, 'specificity determinants,' total hydrophobicity, hydrophobe type and location as design parameters to improve the therapeutic ratio.

Ziqing Jiang; Adriana I. Vasil; Lajos Gera; Michael L. Vasil; Robert S. Hodges

The rapidly growing problem of increased resistance to classical antibiotics makes the development of new classes of antimicrobial agents with lower rates of resistance urgent. Amphipathic cationic α‐helical antimicrobial peptides have been proposed as a potential new class of antimicrobial agents. The goal of this study was to take a broad‐spectrum, 26‐residue, antimicrobial peptide in the all‐D conformation, peptide D1 (K13) with excellent biologic properties and address the question of whether a rational design approach could be used to enhance the biologic properties if the focus was on Gram‐negative pathogens only. To test this hypothesis, we used 11 and 6 diverse strains of Acinetobacter baumannii and Pseudomonas aeruginosa, respectively. We optimized the number and location of positively charged residues on the polar face, the number, location, and type of hydrophobe on the non‐polar face and varied the number of ‘specificity determinants’ in the center of the non‐polar face from 1 to 2 to develop four new antimicrobial peptides. We demonstrated not only improvements in antimicrobial activity, but also dramatic reductions in hemolytic activity and unprecedented improvements in therapeutic indices. Compared to our original starting peptide D1 (V13), peptide D16 had a 746‐fold improvement in hemolytic activity (i.e. decrease), maintained antimicrobial activity, and improved the therapeutic indices by 1305‐fold and 895‐fold against A. baumannii and P. aeruginosa, respectively. The resulting therapeutic indices for D16 were 3355 and 895 for A. baumannii and P. aeruginosa, respectively. D16 is an ideal candidate for commercialization as a clinical therapeutic to treat Gram‐negative bacterial infections.


Protein and Peptide Letters | 2011

Anti-Tuberculosis Activity of α-Helical Antimicrobial Peptides: De Novo Designed L- and D-Enantiomers Versus L- and D-LL37

Ziqing Jiang; Michael Higgins; James Whitehurst; Kevin O. Kisich; Martin I. Voskuil; Robert S. Hodges

With the emergence of multi-drug resistant (MDR) and extensively drug resistant (XDR) Mycobacterium tuberculosis (Mtb), a new class of antimycobacterial agents with very different modes of action compared to classical antibiotics, are urgently needed. In this study, a series of 26-residue, amphipathic, α-helical antimicrobial peptides consisting of all D-amino acid residues and synthetic human L-LL37 (L-enantiomer) and D-LL37 (D-enantiomer) were investigated against M. tuberculosis susceptible strain (H37Rv) and a clinical multi-drug resistant strain (Vertulo). Minimal inhibitory concentrations (MICs) were determined through a peptide killing assay. D5, the most active analog against M. tuberculosis had a MIC value of 11.2 μM (35.2 μg/ml) against H37Rv strain and 15.6 μM (49 μg/ml) against the MDR strain. Peptide D1 had similar activity as D5 against the MDR strain (57 μg/mL), a 9-fold improvement in hemolytic activity and a 7.4-fold better therapeutic index compared to D5. Surprisingly, LL37 enantiomers showed little to no activity compared to the de-novo designed α-helical antimicrobial peptides.


Pharmaceuticals | 2014

“Specificity Determinants” Improve Therapeutic Indices of Two Antimicrobial Peptides Piscidin 1 and Dermaseptin S4 Against the Gram-negative Pathogens Acinetobacter baumannii and Pseudomonas aeruginosa

Ziqing Jiang; Adriana I. Vasil; Michael L. Vasil; Robert S. Hodges

A new class of antimicrobial agents with lower rates of resistance and different targets is urgently needed because of the rapidly increasing resistance to classical antibiotics. Amphipathic cationic α-helical antimicrobial peptides (AMPs) represent such a class of compounds. In our previous studies, using a 26-residue de novo designed antimicrobial peptide, we proposed the concept of “specificity determinant(s)”: positively charged residue(s) in the center of the non-polar face of AMPs that could decrease hemolytic activity/toxicity but increase or maintain the same level of antimicrobial activity to increase dramatically the therapeutic index. In the current study, we used d-enantiomers of two AMPs, Piscidin 1 isolated from fish and dermaseptin S4 isolated from frog. We substituted different positions in the center of the hydrophobic face with one or two lysine residue(s) (one or two “specificity determinant(s)”). This simple modification not only maintained or improved antimicrobial activity against Gram-negative pathogens Acinetobacter baumannii (11 strains) and Pseudomonas aeruginosa (6 strains), but also dramatically decreased hemolytic activity of human red blood cells, as predicted. Therapeutic indices improved by 55-fold and 730-fold for piscidin 1 (I9K) and dermaseptin S4 (L7K, A14K), respectively, against A. baumannii. Similarly, the therapeutic indices improved 32-fold and 980-fold for piscidin 1 (I9K) and dermaseptin S4 (L7K, A14K), respectively, against P. aeruginosa.


Journal of Chromatography A | 2013

An improved approach to hydrophilic interaction chromatography of peptides: Salt gradients in the presence of high isocratic acetonitrile concentrations

Colin T. Mant; Ziqing Jiang; Barry E. Boyes; Robert S. Hodges

Hydrophilic interaction chromatography (HILIC) for separations of peptides has been employed infrequently, particularly considering that this technique was introduced over 20 years ago. The present manuscript describes a radical departure from the traditional HILIC elution approach, where separations are achieved via increasing salt (sodium perchlorate) gradients in the presence of high isocratic concentrations (>80%) of acetonitrile, denoted HILIC/SALT. This initial study compared to reversed-phase chromatography (RPC), HILIC and HILIC/SALT for the separation of mixtures of synthetic peptide standards varying in structure (amphipathic α-helix, random coil), length (10-26 residues), number of positively charged residues (+1 to +11) and hydrophilicity/hydrophobicity. Results showed a marked superiority of the HILIC/SALT approach compared to traditional HILIC and excellent complementarity to RPC for peptide separations. We believe these initial results offer a new dimension to HILIC, enabling it to transform from an occasional HPLC approach for peptide separations to a more generally applicable method.


Biopolymers | 2016

Platform technology to generate broadly cross-reactive antibodies to α-helical epitopes in hemagglutinin proteins from influenza a viruses.

Ziqing Jiang; Lajos Gera; Colin T. Mant; Brooke Hirsch; Zhe Yan; Jonathan A. Shortt; David D. Pollock; Zhaohui Qian; Kathryn V. Holmes; Robert S. Hodges

We have utilized a de novo designed two‐stranded α‐helical coiled‐coil template to display conserved α‐helical epitopes from the stem region of hemagglutinin (HA) glycoproteins of influenza A. The immunogens have all the surface‐exposed residues of the native α‐helix in the native HA protein of interest displayed on the surface of the two‐stranded α‐helical coiled‐coil template. This template when used as an immunogen elicits polyclonal antibodies which bind to the α‐helix in the native protein. We investigated the highly conserved sequence region 421–476 of HA by inserting 21 or 28 residue sequences from this region into our template. The cross‐reactivity of the resulting rabbit polyclonal antibodies prepared to these immunogens was determined using a series of HA proteins from H1N1, H2N2, H3N2, H5N1, H7N7, and H7N9 virus strains which are representative of Group 1 and Group 2 virus subtypes of influenza A. Antibodies from region 449–476 were Group 1 specific. Antibodies to region 421–448 showed the greatest degree of cross‐reactivity to Group 1 and Group 2 and suggested that this region has a great potential as a “universal” synthetic peptide vaccine for influenza A.


Chemical Biology & Drug Design | 2018

Role of positively charged residues on the polar and non-polar faces of amphipathic α-helical antimicrobial peptides on specificity and selectivity for Gram-negative pathogens

Ziqing Jiang; Colin T. Mant; Michael L. Vasil; Robert S. Hodges

We have designed de novo and synthesized eight 26‐residue all D‐conformation amphipathic α‐helical cationic antimicrobial peptides (AMPs), four with “specificity determinants” which provide specificity for prokaryotic cells over eukaryotic cells and four AMPs without specificity determinants. The eight AMPs contain six positively charged Lys residues on the polar face in four different arrangements to understand the role of these residues have on antimicrobial activity against 14 Acinetobacter baumannii strains, seven of which were resistant to polymyxin B and colistin; six diverse Pseudomonas aeruginosa strains and 17 Staphylococcus aureus strains, nine of which were methicillin‐sensitive, and eight of which were methicillin‐resistant. The four AMPs without specificity determinants are extremely hemolytic. In contrast, the four AMPs with specificity determinants had dramatic improvements in therapeutic indices showing the importance of specificity determinants in removing eukaryotic cell toxicity. The specificity determinants combined with the location of positively charged residues on the polar face provide Gram‐negative pathogen selectivity between A. baumannii and S. aureus. Specificity determinants maintain excellent antimicrobial activity in the presence of human sera, whereas the AMPs without specificity determinants were inactive. This study clearly shows the potential of amphipathic α‐helical AMPs with specificity determinants as therapeutics to replace existing antibiotics.


The 24th American Peptide Symposium | 2015

Development of Novel Broad Spectrum Anticancer Small Molecule Peptidomimetics with Nanomolar Activity

Lajos Gera; John J. Tentler; S. Gail Eckhardt; Ziqing Jiang; Angelo D'Alessandro; Robert S. Hodges

Cancer is a major public health problem in the United States and throughout the world. It is currently the second leading cause of death in the United States and is expected to surpass heart diseases in the next few years to become the leading cause of death [1]. The estimated number of new cases of invasive cancer (all types) in the United States is 1,658,370 which is equivalent of more than 4,500 new cancer diagnoses each day. In addition, the estimated number of deaths from cancer in 2015 is 589,430 corresponding to about 1,600 deaths per day [1]. Though there has been a steady increase in survival for most cancers the death rate remains unacceptable and for certain cancers i.e. lung and pancreatic cancers the 5-year relative survival is currently 18% and 7%, respectively. Traditional chemotherapy drugs act against all actively dividing cells (normal and cancerous cells) whereas targeted cancer therapies are drugs that interfere with specific molecular targets involved in cancer cell growth, progression and spread of cancer. Most targeted therapies are either small molecules or monoclonal antibodies. However, therapeutic strategies that target single molecular pathways eventually succumb to problems of intrinsic or acquired resistance due to extensive signaling “cross talk”. Thus, combination targeted therapies are more attractive, as they synergistically inhibit multiple receptors. However, overlapping toxicities and pharmacological interactions limit patient compliance, feasibility and efficacy. Clearly, there is an urgent need to develop new first-line agents with enhanced efficacy and reduced toxicity. We support the concept that the ideal drug maybe a broad spectrum drug whose efficacy is based not on the inhibition of a single target but rather a multi-targeted drug that affects several proteins or events that contribute to the etiology, pathogenesis and progression of diseases [2]. In addition, multipathway targeting is one of the strategies to overcome chemo-resistance. To design novel anticancer drugs with unique structural properties we have taken an innovative and nontraditional approach where we combine pharmacophoric components to create new and highly potent small molecules with a simple three component “A-B-C” structure where each pharmacophore is known to have anticancer properties on its own or when incorporated as a component of an existing drug. Our multi-component “A-B-C” drugs can target simultaneously two or more different molecular targets or molecular mechanisms in a single entity which should reduce the likelihood of drug resistance.


Journal of Chromatography A | 2018

Separation of highly charged (+5 to +10) amphipathic α-helical peptide standards by cation-exchange and reversed-phase high-performance liquid chromatography

Colin T. Mant; Andrew Byars; Sean Ankarlo; Ziqing Jiang; Robert S. Hodges

We are currently examining the potential of amphipathic cationic α-helical peptides as a new generation of peptide standards for both cation-exchange high-performance liquid chromatography and reversed-phase chromatography. Thus, amphipathic peptides are particularly suitable for high-performance liquid chromatography standards due to the preferred binding of the non-polar face to the hydrophobic stationary phase of reversed-phase packings or the preferred binding of the polar face to the charged/hydrophilic stationary phase of cation-exchange packings. The ability of different reversed-phase or cation-exchange matrices to separate mixtures of peptide standards with only subtle hydrophilicity/hydrophobicity variations in both the non-polar and polar face of the peptides can then be assessed. Currently, we have designed de novo a mixture of six 26-residue all D-conformation amphipathic cationic α-helical peptides with a single, positively charged lysine residue in the center of the non-polar face and an increasing number of lysine residues (4-9 residues) replacing neutral residues in the polar face, resulting in an overall net positive charge of +5 to +10. Thus, the non-polar, preferred reversed-phase chromatography binding face remains constant, with only the polar face varying in hydrophilicity/hydrophobicity. Interestingly, even with the non-polar face remaining constant, reversed-phase columns of varying functional group properties (e.g., C8, C18, phenyl, polar endcapped, polar embedded) and porosity (porous versus superficially porous) were able to separate the six peptides in aq. TFA/acetonitrile gradients, albeit with different selectivities. The value of the standards in cation-exchange chromatography was expressed by monitoring the requirement of acetonitrile (0-40% in the mobile phase) to overcome hydrophobic interactions of the peptides with the cation-exchange matrix matrix when eluting with sodium perchlorate gradients at pH 6.5. Interestingly, the resolution of the higher charged peptides (+8,+9,+10) was particularly sensitive to acetonitrile levels. Our results clearly demonstrate the excellent potential of these novel peptide standards to enable optimal column choice and mobile phase conditions for reversed-phase chromatography and cation-exchange chromatography for peptide separations.


The 24th American Peptide Symposium | 2015

Antibody Cross-Reactivity to Hemagglutinin Protein Antigens Demonstrates Feasibility for Development of a “Universal” Influenza A Synthetic Peptide Vaccine

Ziqing Jiang; Lajos Gera; Colin T. Mant; Robert S. Hodges

Influenza A viruses spread rapidly, causing widespread seasonal epidemics of respiratory disease worldwide, which results in more than a billion cases and 500,000 deaths annually [1]. Current influenza vaccines primarily elicit antibodies against the receptor-binding region of the head domain of the hemagglutinin (HA) glycoprotein trimer (Figure 1). This region is hyper-variable and highly mutable, leading to new forms of the virus that can evade neutralizing antibodies. The stem region of HA contains highly conserved α-helical sequences as a result of their functional role in membrane fusion and virus entry. Studies have shown that a few rare neutralizing human monoclonal antibodies can recognize these highly conserved epitopes and neutralize both homotypic and heterotypic influenza strains [2,3]. There are some 18 HA subtypes in influenza A virus that infect humans, animals and birds (Figure 1) [4].

Collaboration


Dive into the Ziqing Jiang's collaboration.

Top Co-Authors

Avatar

Robert S. Hodges

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Colin T. Mant

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Lajos Gera

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Michael L. Vasil

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Adriana I. Vasil

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

James Whitehurst

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Zhaohui Qian

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Zhe Yan

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

John D. Hale

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert E. W. Hancock

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge