1
|
Liu HJ, Li LY, Wang ZL, Fan YL, Shen YX, Song F, Zhu LL. Dynamic polysaccharide/platelet-rich plasma hydrogels with synergistic antibacterial activities for accelerating infected wound healing. Int J Biol Macromol 2024; 281:136209. [PMID: 39383899 DOI: 10.1016/j.ijbiomac.2024.136209] [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: 07/23/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Platelet-rich plasma (PRP) has been recognized as an effective therapy in regenerative medicine and surgery, which can reduce the risk of antibiotic abuse and promote the healing of infected wounds. Recent advances in PRP-based treatments have focused on the controlled release of growth factors in PRP with biocompatible hydrogels and antimicrobial promotion by introducing hydrogel components or antibiotics, while the inherent antimicrobial activity of PRP is mostly neglected or sacrificed. Here, we demonstrate the combination of an antimicrobial polysaccharide, carboxymethyl chitosan, and PRP to construct an antimicrobial hydrogel via dynamic bonding with oxidized chondroitin sulfate. Significant inhibitory effects against Staphylococcus aureus and Escherichia coli (95 % of inhibition rate) are achieved through the synergistic contributions of the polysaccharide and PRP. Additionally, the resulting hydrogel promotes the migration of NIH-3T3 fibroblasts and collagen deposition by approximately 1.7 and 1.8 times, respectively, thereby accelerating the healing process of infected wounds. This work may bring new perspectives for potent applications of PRP-based hydrogel dressings for antibiotic-free management of infected wounds.
Collapse
Affiliation(s)
- Hong-Jie Liu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China; Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550001, China
| | - Lin-Yue Li
- The Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), National Engineering Laboratory of Eco-Friendly Polymeric Materials, (Sichuan), State Key Laboratory of Polymer Materials Engineering, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Zi-Lin Wang
- Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550001, China; Department of Clinical Laboratory, Zigong First People's Hospital, Zigong, Sichuan 643000, China
| | - Ya-Ling Fan
- The Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), National Engineering Laboratory of Eco-Friendly Polymeric Materials, (Sichuan), State Key Laboratory of Polymer Materials Engineering, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yu-Xue Shen
- Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550001, China
| | - Fei Song
- The Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), National Engineering Laboratory of Eco-Friendly Polymeric Materials, (Sichuan), State Key Laboratory of Polymer Materials Engineering, College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Li-Li Zhu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China; Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550001, China.
| |
Collapse
|
2
|
Vasukutty A, Pillarisetti S, Choi J, Kang SH, Park IK. CXCR4 Targeting Nanoplatform for Transcriptional Activation of Latent HIV-1 Infected T Cells. ACS APPLIED BIO MATERIALS 2024; 7:4831-4842. [PMID: 37586084 DOI: 10.1021/acsabm.3c00456] [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: 08/18/2023]
Abstract
Antiretroviral drugs are limited in their ability to target latent retroviral reservoirs in CD4+ T cells, highlighting the need for a T cell-targeted drug delivery system that activates the transcription of inactivated viral DNA in infected cells. Histone deacetylase inhibitors (HDACi) disrupt chromatin-mediated silencing of the viral genome and are explored in HIV latency reversal. But single drug formulations of HDACi are insufficient to elicit therapeutic efficacy, warranting combination therapy. Furthermore, protein kinase C activators (PKC) have shown latency reversal activity in HIV by activating the NF-κB signaling pathway. Combining HDACi (SAHA) with PKC (PMA) activators enhances HIV reservoir activation by promoting chromatin decondensation and subsequent transcriptional activation. In this study, we developed a mixed nanomicelle (PD-CR4) drug delivery system for simultaneous targeting of HIV-infected CD4+ T cells with two drugs, suberoylanilide hydroxamic acid (SAHA) and phorbol 12-myristate 13-acetate (PMA). SAHA is a HDACi that promotes chromatin decondensation, while PMA is a PKC agonist that enhances transcriptional activation. The physicochemical properties of the formulated PD-CR4 nanoparticles were characterized by NMR, CMC, DLS, and TEM analyses. Further, we investigated in vitro safety profiles, targeting efficacy, and transcriptional activation of inactivated HIV reservoir cells. Our results suggest that we successfully prepared a targeted PD system with dual drug loading. We have compared latency reversal efficacy of a single drug nanoformulation and combination drug nanoformulation. Final PD-SP-CR4 successfully activated infected CD4+ T cell reservoirs and showed enhanced antigen release from HIV reservoir T cells, compared with the single drug treatment group as expected. To summarize, our data shows PD-SP-CR4 has potential T cell targeting efficiency and efficiently activated dormant CD4+ T cells. Our data indicate that a dual drug-loaded particle has better therapeutic efficacy than a single loaded particle as expected. Hence, PD-CR4 can be further explored for HIV therapeutic drug delivery studies.
Collapse
Affiliation(s)
- Arathy Vasukutty
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Shin Hyuk Kang
- Departments of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
3
|
Valdivia-Silva J, Chinney-Herrera A. Chemokine receptors and their ligands in breast cancer: The key roles in progression and metastasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:124-161. [PMID: 39260935 DOI: 10.1016/bs.ircmb.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Chemokines and their receptors are a family of chemotactic cytokines with important functions in the immune response in both health and disease. Their known physiological roles such as the regulation of leukocyte trafficking and the development of immune organs generated great interest when it was found that they were also related to the control of early and late inflammatory stages in the tumor microenvironment. In fact, in breast cancer, an imbalance in the synthesis of chemokines and/or in the expression of their receptors was attributed to be involved in the regulation of disease progression, including invasion and metastasis. Research in this area is progressing rapidly and the development of new agents based on chemokine and chemokine receptor antagonists are emerging as attractive alternative strategies. This chapter provides a snapshot of the different functions reported for chemokines and their receptors with respect to the potential to regulate breast cancer progression.
Collapse
Affiliation(s)
- Julio Valdivia-Silva
- Centro de Investigación en Bioingenieria (BIO), Universidad de Ingenieria y Tecnologia-UTEC, Barranco, Lima, Peru.
| | - Alberto Chinney-Herrera
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico-UNAM, Ciudad Universitaria, Coyoacan, Ciudad de Mexico, Mexico
| |
Collapse
|
4
|
Pampalone M, Cuscino N, Iannolo G, Amico G, Ricordi C, Vitale G, Carcione C, Castelbuono S, Scilabra SD, Coronnello C, Gruttadauria S, Pietrosi G. Human Amniotic MSC Response in LPS-Stimulated Ascites from Patients with Cirrhosis: FOXO1 Gene and Th17 Activation in Enhanced Antibacterial Activation. Int J Mol Sci 2024; 25:2801. [PMID: 38474048 DOI: 10.3390/ijms25052801] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Spontaneous bacterial peritonitis (SBP) is a severe complication in patients with decompensated liver cirrhosis and is commonly treated with broad spectrum antibiotics. However, the rise of antibiotic resistance requires alternative therapeutic strategies. As recently shown, human amnion-derived mesenchymal stem cells (hA-MSCs) are able, in vitro, to promote bacterial clearance and modulate the immune and inflammatory response in SBP. Our results highlight the upregulation of FOXO1, CXCL5, CXCL6, CCL20, and MAPK13 in hA-MSCs as well as the promotion of bacterial clearance, prompting a shift in the immune response toward a Th17 lymphocyte phenotype after 72 h treatment. In this study, we used an in vitro SBP model and employed omics techniques (next-generation sequencing) to investigate the mechanisms by which hA-MSCs modify the crosstalk between immune cells in LPS-stimulated ascitic fluid. We also validated the data obtained via qRT-PCR, cytofluorimetric analysis, and Luminex assay. These findings provide further support to the hope of using hA-MSCs for the prevention and treatment of infective diseases, such as SBP, offering a viable alternative to antibiotic therapy.
Collapse
Affiliation(s)
- Mariangela Pampalone
- Ri.MED Foundation, 90127 Palermo, Italy
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Nicola Cuscino
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Gioacchin Iannolo
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giandomenico Amico
- Ri.MED Foundation, 90127 Palermo, Italy
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA
| | | | | | - Salvatore Castelbuono
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Simone Dario Scilabra
- Ri.MED Foundation, 90127 Palermo, Italy
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | | | - Salvatore Gruttadauria
- Department for the Treatment and Study of Abdominal Disease and Abdominal Transplantation, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), UPMCI (University of Pittsburgh Medical Center Italy), 90127 Palermo, Italy
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95124 Catania, Italy
| | - Giada Pietrosi
- Department for the Treatment and Study of Abdominal Disease and Abdominal Transplantation, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), UPMCI (University of Pittsburgh Medical Center Italy), 90127 Palermo, Italy
| |
Collapse
|
5
|
Blanchet X, Weber C, von Hundelshausen P. Chemokine Heteromers and Their Impact on Cellular Function-A Conceptual Framework. Int J Mol Sci 2023; 24:10925. [PMID: 37446102 DOI: 10.3390/ijms241310925] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Chemoattractant cytokines or chemokines are proteins involved in numerous biological activities. Their essential role consists of the formation of gradient and (immune) cell recruitment. Chemokine biology and its related signaling system is more complex than simple ligand-receptor interactions. Beside interactions with their cognate and/or atypical chemokine receptors, and glycosaminoglycans (GAGs), chemokines form complexes with themselves as homo-oligomers, heteromers and also with other soluble effector proteins, including the atypical chemokine MIF, carbohydrate-binding proteins (galectins), damage-associated molecular patterns (DAMPs) or with chemokine-binding proteins such as evasins. Likewise, nucleic acids have been described as binding targets for the tetrameric form of CXCL4. The dynamic balance between monomeric and dimeric structures, as well as interactions with GAGs, modulate the concentrations of free chemokines available along with the nature of the gradient. Dimerization of chemokines changes the canonical monomeric fold into two main dimeric structures, namely CC- and CXC-type dimers. Recent studies highlighted that chemokine dimer formation is a frequent event that could occur under pathophysiological conditions. The structural changes dictated by chemokine dimerization confer additional biological activities, e.g., biased signaling. The present review will provide a short overview of the known functionality of chemokines together with the consequences of the interactions engaged by the chemokines with other proteins. Finally, we will present potential therapeutic tools targeting the chemokine multimeric structures that could modulate their biological functions.
Collapse
Affiliation(s)
- Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| |
Collapse
|
6
|
Souza E Silva P, Ferreira MA, de Moraes LFR, de Barros E, Preza SLE, Cardoso MH, Franco OL, Migliolo L. Synthetic peptides bioinspired in temporin-PTa with antibacterial and antibiofilm activity. Chem Biol Drug Des 2022; 100:51-63. [PMID: 35377553 DOI: 10.1111/cbdd.14052] [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: 10/18/2021] [Revised: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 12/25/2022]
Abstract
Several antimicrobial peptides (AMPs) have been reported in amphibian toxins, as temporin-PTa from Hylarana picturata. The amino acid distribution within a helical structure of AMPs favors the design of new bioactive peptides. Therefore, this work reports the rational design of two new synthetic peptides denominated Hp-MAP1 and Hp-MAP2 derived from temporin-PTa. These peptides present an amphipathic helix with positive charges of +4 and +5, hydrophobic moment (<µH>) of 0.66 and 0.72 and hydrophobicity (<H>) of 0.49 and 0.41, respectively. Hp-MAP1 and Hp-MAP2 displayed in vitro activity against Gram-negative and Gram-positive bacteria from 2.8 to 92 µM, without presenting hemolytic effects. Molecular dynamics simulation suggested that the parent and designed temporin-like peptides lack structural stability in an aqueous solution. By contrast, α-helical structures were predicted in hydrophobic and anionic environments. Additionally, the peptides were simulated on mimetic membranes composed of anionic and neutral phospholipids 1,2-dipalmitoylsn-glycerol-3-phosphatidylglycerol (DPPG-anionic), 1,2-dipalmitoyl-sn-lyco-3 phosphatidylethanolamine (DPPE-neutral). When in contact with DPPG/DPPE (90:10) and DPPG/DPPE (50:50) temporin-PTa, Hp-MAP1 and Hp-MAP2 established interactions guided by hydrogen and saline bounds. Therefore, the findings described here reveal that the optimization of the amphipathic α-helical cationic peptides Hp-MAP1 and Hp-MAP2 enabled the generation of new synthetic antimicrobial agents to combat pathogenic microorganisms.
Collapse
Affiliation(s)
- Patrícia Souza E Silva
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Marcos Antonio Ferreira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil
| | | | - Elizângela de Barros
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | | | - Marlon Henrique Cardoso
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Programa de Pós-Graduação em Biotecnologia e Ciências Genômicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Octávio Luiz Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Programa de Pós-Graduação em Biotecnologia e Ciências Genômicas, Universidade Católica de Brasília, Brasília, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Ludovico Migliolo
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil.,Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
7
|
Yang X, Wu Y, Zhang P, Chen G, Cao Z, Ao J, Sun Y, Zhou Y. CC chemokine 1 protein from Cromileptes altivelis (CaCC1) promotes antimicrobial immune defense. FISH & SHELLFISH IMMUNOLOGY 2022; 123:102-112. [PMID: 35240293 DOI: 10.1016/j.fsi.2022.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Chemokines are a family of small signaling proteins that are secreted by various cells. In addition to their roles in immune surveillance, localization of antigen, and lymphocyte trafficking for the maintenance of homeostasis, chemokines also function in induce immune cell migration under pathological conditions. In the present study, a novel CC chemokine gene (CaCC1) from humpback grouper (Cromileptes altivelis) was cloned and characterized. CaCC1 comprised a 435 bp open reading frame encoding 144 amino acid residues. The putative molecular weight of CaCC1 protein was 15 kDa CaCC1 contains four characteristic cysteines that are conserved in other known CC chemokines. CaCC1 also shares 11.64%-90.28% identity with other teleost and mammal CC chemokines. Phylogenetic analysis revealed that CaCC1 is most closely related to Epinephelus coioides EcCC1, both of which are in a fish-specific CC chemokine clade. CaCC1 was constitutively expressed in all examined C. altivelis tissues, with high expression levels in skin, heart, liver, and intestine. Vibrio harveyi stimulation up-regulated CaCC1 expression levels in liver, spleen, and head-kidney. Functional analyses revealed that the recombinant protein (rCaCC1) could induce the migration of head-kidney lymphocytes from C. altivelis. Moreover, rCaCC1 significantly enhanced phagocytosis in head-kidney macrophages from C. altivelis. In addition, rCaCC1 exhibited antimicrobial activities against Staphylococcus aureus, Edwardsiella tarda, and V. harveyi. In vivo, CaCC1 overexpression improved bacterial clearance in V. harveyi infected fish. Conversely, CaCC1 knockdown resulted in a significant decrease of bacterial clearance. These results demonstrate the important roles that CaCC1 plays in homeostasis and in inflammatory response to bacterial infection.
Collapse
Affiliation(s)
- Xiaoyu Yang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, PR China
| | - Ying Wu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China
| | - Panpan Zhang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, PR China
| | - Guisen Chen
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China
| | - Zhenjie Cao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China
| | - Jingqun Ao
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, PR China
| | - Yun Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, PR China.
| | - Yongcan Zhou
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, PR China.
| |
Collapse
|
8
|
Davidson L, van den Reek JM, Bruno M, van Hunsel F, Herings RM, Matzaraki V, Boahen CK, Kumar V, Groenewoud HM, van de Veerdonk FL, Netea MG, de Jong EM, Kullberg BJ. Risk of candidiasis associated with interleukin-17 inhibitors: A real-world observational study of multiple independent sources. THE LANCET REGIONAL HEALTH. EUROPE 2022; 13:100266. [PMID: 34950923 PMCID: PMC8671639 DOI: 10.1016/j.lanepe.2021.100266] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Biologics directed against the T-helper (Th)-17 pathway have been approved for several inflammatory diseases. Interleukin (IL)-17 is involved in anti- Candida host defense, and clinical trials suggested increased candidiasis incidence during IL-17 inhibitor therapy. We describe the worldwide epidemiology of candidiasis during Th17 inhibitor therapy, and immunological mechanisms involved in candidiasis susceptibility. METHODS A comprehensive analysis of multiple independent sources reporting Candida adverse events during biologics inhibiting the Th17 pathway was performed. Association between Th17 inhibitors and candidiasis was assessed using safety reports of (1) WHO and (2) EMA, (3) a population-based prescriptions registry, and (4) a psoriasis cohort. In a cohort of psoriasis patients experiencing candidiasis during Th17 inhibitors, Candida killing by immune cells and serum inflammatory proteome were analyzed. FINDINGS A strong association between IL-17 inhibitors and candidiasis (ROR 10·20) was found in the WHO database, particularly for cutaneous (ROR 12·28), oropharyngeal (ROR 19·18), and esophageal candidiasis (ROR 21·20). Risk was higher relative to TNF-α inhibitors (4-10-fold, depending on candidiasis type), confirmed by EMA reports (16-33-fold), prescriptions registry (2-42-fold), and a psoriasis cohort (3-25-fold). After start of IL-17 inhibitors, patients' risk of candidiasis requiring antifungals increased 2-16 fold. In the psoriasis cohort, 58% of IL-17 treatment episodes were associated with candidiasis. In Th17 inhibitor recipients, proteins involved in anti- Candida immunity and Candida killing by mononuclear leukocytes were impaired. INTERPRETATION IL-17 inhibitors are associated with an increased risk of oropharyngeal, esophageal, and cutaneous candidiasis, posing a significant disease burden for IL-17 inhibitor recipients. FUNDING RadboudUMC.
Collapse
Affiliation(s)
- Linda Davidson
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Mariolina Bruno
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Florence van Hunsel
- Netherlands Pharmacovigilance Centre Lareb, ‘s Hertogenbosch, the Netherlands
| | - Ron M.C. Herings
- PHARMO Institute for Drug Outcomes Research, Utrecht, the Netherlands
- Department of Epidemiology & Data Science, Amsterdam UMC – Vrije Universiteit, Amsterdam Public Health, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Collins K. Boahen
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vinod Kumar
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans M.M. Groenewoud
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Elke M.G.J. de Jong
- Department of Dermatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bart Jan Kullberg
- Department of Internal Medicine and Radboudumc Center for Infectious diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
9
|
Shoari A, Khodabakhsh F, Ahangari Cohan R, Salimian M, Karami E. Anti-angiogenic peptides application in cancer therapy; a review. Res Pharm Sci 2021; 16:559-574. [PMID: 34760005 PMCID: PMC8562409 DOI: 10.4103/1735-5362.327503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/16/2021] [Accepted: 09/19/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer is a disease advanced via surplus angiogenesis. The development of new anti-angiogenic therapeutic agents with more efficacy and fewer side effects is still quite necessary. Conventional therapies saving the life of many cancer patients but due to drug resistance and lack of specificity utilizing these methods is faced with limits. Recently, new therapeutic agents have been developed and used to treat cancers such as scaffold proteins, monoclonal antibodies, tyrosine kinase inhibitors, and peptides. In antiangiogenic drug development, anti-angiogenic peptides design is a significant aim. Peptides have developed as substantial therapeutics that are being carefully investigated in angiogenesis-dependent diseases because of their high penetrating rate into the cancer cells, high specificity, and low toxicity. In this review, we focus on anti-angiogenic peptides in the field of cancer therapy that are designed, screened, or derived from nanobodies, mimotopes, phage displays, and natural resources.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, I.R. Iran
| | - Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, I.R. Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, I.R. Iran
| | - Morteza Salimian
- Department of Medical Laboratory, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Elmira Karami
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, I.R. Iran
| |
Collapse
|
10
|
Konger RL, Derr-Yellin E, Zimmers TA, Katona T, Xuei X, Liu Y, Zhou HM, Simpson ER, Turner MJ. Epidermal PPARγ Is a Key Homeostatic Regulator of Cutaneous Inflammation and Barrier Function in Mouse Skin. Int J Mol Sci 2021; 22:ijms22168634. [PMID: 34445339 PMCID: PMC8395473 DOI: 10.3390/ijms22168634] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 01/10/2023] Open
Abstract
Both agonist studies and loss-of-function models indicate that PPARγ plays an important role in cutaneous biology. Since PPARγ has a high level of basal activity, we hypothesized that epidermal PPARγ would regulate normal homeostatic processes within the epidermis. In this current study, we performed mRNA sequencing and differential expression analysis of epidermal scrapings from knockout mice and wildtype littermates. Pparg-/-epi mice exhibited a 1.5-fold or greater change in the expression of 11.8% of 14,482 identified transcripts. Up-regulated transcripts included those for a large number of cytokines/chemokines and their receptors, as well as genes associated with inflammasome activation and keratinization. Several of the most dramatically up-regulated pro-inflammatory genes in Pparg-/-epi mouse skin included Igfl3, 2610528A11Rik, and Il1f6. RT-PCR was performed from RNA obtained from non-lesional full-thickness skin and verified a marked increase in these transcripts, as well as transcripts for Igflr1, which encodes the receptor for Igfl3, and the 2610528A11Rik receptor (Gpr15). Transcripts for Il4 were detected in Pparg-/-epi mouse skin, but transcripts for Il17 and Il22 were not detected. Down-regulated transcripts included sebaceous gland markers and a number of genes associated with lipid barrier formation. The change in these transcripts correlates with an asebia phenotype, increased transepidermal water loss, alopecia, dandruff, and the appearance of spontaneous inflammatory skin lesions. Histologically, non-lesional skin showed hyperkeratosis, while inflammatory lesions were characterized by dermal inflammation and epidermal acanthosis, spongiosis, and parakeratosis. In conclusion, loss of epidermal Pparg alters a substantial set of genes that are associated with cutaneous inflammation, keratinization, and sebaceous gland function. The data indicate that epidermal PPARγ plays an important role in homeostatic epidermal function, particularly epidermal differentiation, barrier function, sebaceous gland development and function, and inflammatory signaling.
Collapse
Affiliation(s)
- Raymond L. Konger
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Correspondence: ; Tel.: +1-317-274-4154
| | - Ethel Derr-Yellin
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Terrence Katona
- Department of Pathology & Laboratory Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; (E.D.-Y.); (T.K.)
| | - Xiaoling Xuei
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Yunlong Liu
- The Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (T.A.Z.); (Y.L.)
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Hong-Ming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
| | - Ed Ronald Simpson
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew J. Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.-M.Z.); (M.J.T.)
- Department of Dermatology, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
11
|
Sanegre S, Eritja N, de Andrea C, Diaz-Martin J, Diaz-Lagares Á, Jácome MA, Salguero-Aranda C, García Ros D, Davidson B, Lopez R, Melero I, Navarro S, Ramon Y Cajal S, de Alava E, Matias-Guiu X, Noguera R. Characterizing the Invasive Tumor Front of Aggressive Uterine Adenocarcinoma and Leiomyosarcoma. Front Cell Dev Biol 2021; 9:670185. [PMID: 34150764 PMCID: PMC8209546 DOI: 10.3389/fcell.2021.670185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
The invasive tumor front (the tumor–host interface) is vitally important in malignant cell progression and metastasis. Tumor cell interactions with resident and infiltrating host cells and with the surrounding extracellular matrix and secreted factors ultimately determine the fate of the tumor. Herein we focus on the invasive tumor front, making an in-depth characterization of reticular fiber scaffolding, infiltrating immune cells, gene expression, and epigenetic profiles of classified aggressive primary uterine adenocarcinomas (24 patients) and leiomyosarcomas (11 patients). Sections of formalin-fixed samples before and after microdissection were scanned and studied. Reticular fiber architecture and immune cell infiltration were analyzed by automatized algorithms in colocalized regions of interest. Despite morphometric resemblance between reticular fibers and high presence of macrophages, we found some variance in other immune cell populations and distinctive gene expression and cell adhesion-related methylation signatures. Although no evident overall differences in immune response were detected at the gene expression and methylation level, impaired antimicrobial humoral response might be involved in uterine leiomyosarcoma spread. Similarities found at the invasive tumor front of uterine adenocarcinomas and leiomyosarcomas could facilitate the use of common biomarkers and therapies. Furthermore, molecular and architectural characterization of the invasive front of uterine malignancies may provide additional prognostic information beyond established prognostic factors.
Collapse
Affiliation(s)
- Sabina Sanegre
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| | - Núria Eritja
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Carlos de Andrea
- Cancer CIBER (CIBERONC), Madrid, Spain.,Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Juan Diaz-Martin
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Ángel Diaz-Lagares
- Cancer CIBER (CIBERONC), Madrid, Spain.,Cancer Epigenomics, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - María Amalia Jácome
- Department of Mathematics, MODES Group, CITIC, Faculty of Science, Universidade da Coruña, A Coruña, Spain
| | - Carmen Salguero-Aranda
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - David García Ros
- Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Ben Davidson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Rafel Lopez
- Cancer CIBER (CIBERONC), Madrid, Spain.,Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.,Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Ignacio Melero
- Cancer CIBER (CIBERONC), Madrid, Spain.,Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Samuel Navarro
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| | - Santiago Ramon Y Cajal
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Enrique de Alava
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institute of Biomedicine of Sevilla, Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Xavier Matias-Guiu
- Cancer CIBER (CIBERONC), Madrid, Spain.,Institut de Recerca Biomèdica de LLeida (IRBLLEIDA), Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Department of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, University of Lleida - University of Barcelona, Barcelona, Spain
| | - Rosa Noguera
- Cancer CIBER (CIBERONC), Madrid, Spain.,Department of Pathology, School of Medical, University of Valencia-INCLIVA, Valencia, Spain
| |
Collapse
|
12
|
Li L, Zhang L, Zhang T, Qi X, Cheng G, Xia L. Serum Chemokine CXCL7 as a Potential Novel Biomarker for Obstructive Colorectal Cancer. Front Oncol 2021; 10:599363. [PMID: 33643903 PMCID: PMC7902867 DOI: 10.3389/fonc.2020.599363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/07/2020] [Indexed: 01/10/2023] Open
Abstract
Due to the lack of typical symptoms and signs and sensitive indicators for early diagnosis of obstructive colorectal cancer (OCRC), it is critically needed to find new novel biomarkers to ameliorate the management of OCRC patients. In this study, 472 blood samples were collected and measured by enzyme-linked immunosorbent assay (ELISA) to investigate the value of serum chemokine ligand 7 (CXCL7) in diagnosis and prognosis for OCRC patients. The median concentrations of CXCL7 in non-OCRC and OCRC were both higher than that in controls (both P < 0.05). Importantly, the median serum concentration of CXCL7 in OCRC was also higher than that in non-OCRC (P < 0.001). In all OCRC patients, the area under the curve (AUC) of CXCL7 was 0.918 with a sensitivity of 86.54% and a specificity of 81.87%. Similarly, the AUC of CXCL7 was 0.684 when the diagnostic test was performed between OCRC and CRC patients. CXCL7 had a higher AUC than other markers. The concentration of CXCL7 in 40 postoperative OCRC patients was higher than normal people and lower than preoperative patients. The median survival time was 62.00 months and the 5-year overall survival (OS) rate of the patients was 51.80% in all 155 OCRC patients. Multivariate Cox proportional hazard regression model analysis showed that high CXCL7 in serum was independent factors associated with poor OS of OCRC patients (HR = 2.216, P = 0.032). These results demonstrate that serum CXCL7 may be a potential biomarker both in diagnosis and prognosis for OCRC patients.
Collapse
Affiliation(s)
- Longhai Li
- Department of Science and Education, The People's Hospital of Bozhou, Bozhou, China
| | - Lihua Zhang
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ting Zhang
- Department of Pathology Cancer Research Center, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaowei Qi
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Gang Cheng
- Department of Oncology, The People's Hospital of Bozhou, Bozhou, China
| | - Lingxia Xia
- Department of Science and Education, The People's Hospital of Bozhou, Bozhou, China
| |
Collapse
|
13
|
Common and Differential Dynamics of the Function of Peripheral Blood Mononuclear Cells between Holstein and Jersey Cows in Heat-Stress Environment. Animals (Basel) 2020; 11:ani11010019. [PMID: 33374309 PMCID: PMC7824059 DOI: 10.3390/ani11010019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Seasonal change, particularly changing to hot and humid season, has a negative effect on dairy cows in various ways, including productivity, reproduction, metabolism, and immunity. In high-temperature and humid weather, dairy cows are vulnerable to diseases by weakened immune system. However, the cause of this has not been fully described. Therefore, this study aims to understand changes of specific gene expression and immune pathways based on transcriptome analysis from peripheral blood mononuclear cells of Holstein and Jersey dairy cows between normal and heat-stress environmental conditions. We observed that the two breeds of dairy cow have common and different immune shifts according to the changes of temperature and humidity condition. Overall, the findings of this study improve the understanding of the underlying mechanisms by which seasonal changes affect dairy cow immunity. Abstract Heat stress has been reported to affect the immunity of dairy cows. However, the mechanisms through which this occurs are not fully understood. Two breeds of dairy cow, Holstein and Jersey, have distinct characteristics, including productivity, heat resistance, and disease in high-temperature environments. The objective of this study is to understand the dynamics of the immune response of two breeds of dairy cow to environmental change. Ribonucleic acid sequencing (RNA-seq) results were analyzed to characterize the gene expression change of peripheral blood mononuclear cells (PBMCs) in Holstein and Jersey cows between moderate temperature-humidity index (THI) and high THI environmental conditions. Many of the differentially expressed genes (DEGs) identified are associated with critical immunological functions, particularly phagocytosis, chemokines, and cytokine response. Among the DEGs, CXCL3 and IL1A were the top down-regulated genes in both breeds of dairy cow, and many DEGs were related to antimicrobial immunity. Functional analysis revealed that cytokine and chemokine response-associated pathways in both Holstein and Jersey PBMCs were the most important pathways affected by the THI environmental condition. However, there were also breed-specific genes and pathways that altered according to THI environmental condition. Collectively, there were both common and breed-specific altered genes and pathways in Holstein and Jersey cows. The findings of this study expand our understanding of the dynamics of immunity in different breeds of dairy cow between moderate THI and high THI environmental conditions.
Collapse
|
14
|
Interleukin 34 Serves as a Novel Molecular Adjuvant against Nocardia Seriolae Infection in Largemouth Bass ( Micropterus Salmoides). Vaccines (Basel) 2020; 8:vaccines8020151. [PMID: 32231137 PMCID: PMC7349345 DOI: 10.3390/vaccines8020151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
DNA vaccines have been widely employed in controlling viral and bacterial infections in mammals and teleost fish. Co-injection of molecular adjuvants, including chemokines, cytokines, and immune co-stimulatory molecules, is one of the potential strategies used to improve DNA vaccine efficacy. In mammals and teleost fish, interleukin-34 (IL-34) had been described as a multifunctional cytokine and its immunological role had been confirmed; however, the adjuvant capacity of IL-34 remains to be elucidated. In this study, IL-34 was identified in largemouth bass. A recombinant plasmid of IL-34 (pcIL-34) was constructed and co-administered with a DNA vaccine encoding hypoxic response protein 1 (Hrp1; pcHrp1) to evaluate the adjuvant capacity of pcIL-34 against Nocardia seriolae infection. Our results indicated that pcIL-34 co-injected with pcHrp1 not only triggered innate immunity and a specific antibody response, but also enhanced the mRNA expression level of immune-related genes encoding for cytokines, chemokines, and humoral and cell-mediated immunity. Moreover, pcIL-34 enhanced the protection of pcHrp1 against N. seriolae challenge and conferred the relative percent survival of 82.14%. Collectively, IL-34 is a promising adjuvant in a DNA vaccine against nocardiosis in fish.
Collapse
|
15
|
Zhang W, Guo Y, Kuss M, Shi W, Aldrich AL, Untrauer J, Kielian T, Duan B. Platelet-Rich Plasma for the Treatment of Tissue Infection: Preparation and Clinical Evaluation. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:225-236. [PMID: 30712506 DOI: 10.1089/ten.teb.2018.0309] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPACT STATEMENT The clinical application of platelet-rich plasma (PRP) has been widely studied for its effects on trauma or injury repair/regeneration, however the antibacterial property of PRP has been overlooked. Increasing evidence suggests PRP as a good antibacterial agent and that it could help prevent/treat tissue infection. This review emphasizes the importance of PRP's antibacterial property and summarizes the preclinical and clinical findings regarding the application of PRP in the prevention and treatment of wound and bone infection. The use of biocompatible PRP may be advantageous for tissue infection treatment due to its inherent antibacterial and healing promoting properties.
Collapse
Affiliation(s)
- Wenhai Zhang
- 1 Department of Orthopedics, Tianjin Hospital, Tianjin, People's Republic of China.,2 Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yue Guo
- 3 Tissue Engineering Labs of Orthopedics Institute, Tianjin Hospital, Tianjin, People's Republic of China
| | - Mitchell Kuss
- 2 Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,4 Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Wen Shi
- 2 Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,4 Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Amy L Aldrich
- 5 Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jason Untrauer
- 6 Division of Oral and Maxillofacial Surgery, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Tammy Kielian
- 5 Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Bin Duan
- 2 Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,4 Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,7 Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,8 Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
16
|
Crawford MA, Margulieux KR, Singh A, Nakamoto RK, Hughes MA. Mechanistic insights and therapeutic opportunities of antimicrobial chemokines. Semin Cell Dev Biol 2019; 88:119-128. [PMID: 29432954 PMCID: PMC6613794 DOI: 10.1016/j.semcdb.2018.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/06/2018] [Indexed: 12/27/2022]
Abstract
Chemokines are a family of small proteins best known for their ability to orchestrate immune cell trafficking and recruitment to sites of infection. Their role in promoting host defense is multiplied by a number of additional receptor-dependent biological activities, and most, but not all, chemokines have been found to mediate direct antimicrobial effects against a broad range of microorganisms. The molecular mechanism(s) by which antimicrobial chemokines kill bacteria remains unknown; however, recent observations have expanded our fundamental understanding of chemokine-mediated bactericidal activity to reveal increasingly diverse and complex actions. In the current review, we present and consider mechanistic insights of chemokine-mediated antimicrobial activity against bacteria. We also discuss how contemporary advances are reshaping traditional paradigms and opening up new and innovative avenues of research with translational implications. Towards this end, we highlight a developing framework for leveraging chemokine-mediated bactericidal and immunomodulatory effects to advance pioneering therapeutic approaches for treating bacterial infections, including those caused by multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Matthew A Crawford
- Division of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Katie R Margulieux
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Arpita Singh
- Division of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Robert K Nakamoto
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Molly A Hughes
- Division of Infectious Diseases & International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
17
|
Therapeutic strategies to target microbial protein-glycosaminoglycan interactions. Biochem Soc Trans 2018; 46:1505-1515. [PMID: 30381333 DOI: 10.1042/bst20170485] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 09/06/2018] [Accepted: 09/18/2018] [Indexed: 01/03/2023]
Abstract
Glycans are involved in a plethora of human pathologies including infectious diseases. Especially, glycosaminoglycans (GAGs), like heparan sulfate and chondroitin sulfate, have been found to be involved in different crucial stages of microbial invasion. Here, we review various therapeutic approaches, which target the interface of host GAGs and microbial proteins and discuss their limitations and challenges for drug development.
Collapse
|
18
|
Targeting luteinizing hormone-releasing hormone: A potential therapeutics to treat gynecological and other cancers. J Control Release 2018; 269:277-301. [DOI: 10.1016/j.jconrel.2016.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 01/05/2023]
|
19
|
Nazari A, Khorramdelazad H, Hassanshahi G. Biological/pathological functions of the CXCL12/CXCR4/CXCR7 axes in the pathogenesis of bladder cancer. Int J Clin Oncol 2017; 22:991-1000. [PMID: 29022185 DOI: 10.1007/s10147-017-1187-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022]
Abstract
CXC chemokine ligand 12 (CXCL12) is an important member of the CXC subfamily of chemokines, and has been extensively studied in various human body organs and systems, both in physiological and clinical states. Ligation of CXCL12 to CXCR4 and CXCR7 as its receptors on peripheral immune cells gives rise to pleiotropic activities. CXCL12 itself is a highly effective chemoattractant which conservatively attracts lymphocytes and monocytes, whereas there exists no evidence to show attraction for neutrophils. CXCL12 regulates inflammation, neo-vascularization, metastasis, and tumor growth, phenomena which are all pivotally involved in cancer development and further metastasis. Generation and secretion of CXCL12 by stromal cells facilitate attraction of cancer cells, acting through its cognate receptor, CXCR4, which is expressed by both hematopoietic and non-hematopoietic tumor cells. CXCR4 stimulates tumor progression by different mechanisms and is required for metastatic spread to organs where CXCL12 is expressed, thereby allowing tumor cells to access cellular niches, such as the marrow, which favor tumor cell survival and proliferation. It has also been demonstrated that CXCL12 binds to another seven-transmembrane G-protein receptor or G-protein-coupled receptor, namely CXCR7. These studies indicated critical roles for CXCR4 and CXCR7 mediation of tumor metastasis in several types of cancers, suggesting their contributions as biomarkers of tumor behavior as well as potential therapeutic targets. Furthermore, CXCL12 itself has the capability to stimulate survival and growth of neoplastic cells in a paracrine fashion. CXCL12 is a supportive chemokine for tumor neovascularization via attracting endothelial cells to the tumor microenvironment. It has been suggested that elevated protein and mRNA levels of CXCL12/CXCR4/CXCR7 are associated with human bladder cancer (BC). Taken together, mounting evidence suggests a role for CXCR4, CXCR7, and their ligand CXCL12 during the genesis of BC and its further development. However, a better understanding is still required before exploring CXCL12/CXCR4/CXCR7 targeting in the clinic.
Collapse
Affiliation(s)
- Alireza Nazari
- Department of Surgery, School of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran.,Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran. .,Department of Immunology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
20
|
Patel S, Akhtar N. Antimicrobial peptides (AMPs): The quintessential 'offense and defense' molecules are more than antimicrobials. Biomed Pharmacother 2017; 95:1276-1283. [PMID: 28938518 DOI: 10.1016/j.biopha.2017.09.042] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial peptides (AMPs) are cationic amphiphilic molecules with α-helix or β-sheet linear motifs and linear or cyclic configurations. For their role in 'defense and offense', they are present in all living organisms. AMPs are named so, as they inhibit a wide array of microbes by membrane pore formation and subsequent perturbation of mitochondrial membrane ionic balance. However, their functional repertoire is expanding with validated roles in cytotoxicity, wound healing, angiogenesis, apoptosis, and chemotaxis [1]. A number of endogenous AMPs have been characterized in human body such as defensins, cathelicidins, histatins etc. They mediate critical functions, but when homeostasis is broken, they turn hostile and initiate inflammatory diseases. This review discusses the sources of therapeutic AMPs; auto-immunity risks of endogenous AMPs, and their dermatological applications; normally overlooked risks of the peptides; and scopes ahead. This holistic work is expected to be a valuable reference for further research in this field.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego 92182, USA.
| | - Nadeem Akhtar
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
21
|
Cossio-Ayala M, Domínguez-López M, Mendez-Enriquez E, Portillo-Téllez MDC, García-Hernández E. In vitro and in vivo antimicrobial activity of a synthetic peptide derived from the C-terminal region of human chemokine CCL13 against Pseudomonas aeruginosa. Peptides 2017; 94:49-55. [PMID: 28687455 DOI: 10.1016/j.peptides.2017.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/26/2017] [Accepted: 06/26/2017] [Indexed: 01/12/2023]
Abstract
Chemokines are important mediators of immunological responses during inflammation and under steady-state conditions. In addition to regulating cell migration, some chemotactic cytokines have direct effects on bacteria. Here, we characterized the antibacterial ability of the synthetic oligopeptide CCL1357-75, which corresponds to the carboxyl-terminal region of the human chemokine CCL13. In vitro measurements indicated that CCL1357-75 disrupts the cell membrane of Pseudomonas aeruginosa through a mechanism coupled to an unordered-helicoidal conformational transition. In a murine pneumonic model, CCL1357-75 improved mouse survival and bacterial clearance and decreased neutrophil recruitment, proinflammatory cytokines and lung pathology compared with that observed in untreated infected animals. Overall, our study supports the ability of chemokines and/or chemokine-derived oligopeptides to act as direct defense agents against pathogenic bacteria and suggests their potential use as alternative antibiotics.
Collapse
Affiliation(s)
- Mayte Cossio-Ayala
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04630, D.F., Mexico; Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04630, D.F., Mexico
| | - Mariana Domínguez-López
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04630, D.F., Mexico
| | - Erika Mendez-Enriquez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04630, D.F., Mexico
| | | | - Enrique García-Hernández
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04630, D.F., Mexico.
| |
Collapse
|
22
|
Short peptides interfering with signaling pathways as new therapeutic tools for cancer treatment. Future Med Chem 2017; 9:199-221. [PMID: 28111982 DOI: 10.4155/fmc-2016-0189] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Short peptides have many advantages, such as low molecular weight, selectivity for a specific target, organelles or cells with minimal toxicity. We describe properties of short peptides, which interfere with communication networks in tumor cells and within microenvironment of malignant gliomas, the most common brain tumors. We focus on ligand/receptor axes and intracellular signaling pathways critical for gliomagenesis that could be targeted with interfering peptides. We review structures and efficacy of organelle-specific and cell-penetrating peptides and describe diverse chemical modifications increasing proteolytic stability and protecting synthetic peptides against degradation. We report results of application of short peptides in glioma therapy clinical trials, their rises and falls. The most advanced examples of therapeutics such as short interfering peptides combined with cell-penetrating peptides that show good effectiveness in disease models are presented. It is foreseen that identification of peptides with better clinical properties may improve their success rates in clinical trials.
Collapse
|
23
|
Vasilchenko AS, Rogozhin EA, Vasilchenko AV, Kartashova OL, Sycheva MV. Novel haemoglobin-derived antimicrobial peptides from chicken (Gallus gallus) blood: purification, structural aspects and biological activity. J Appl Microbiol 2016; 121:1546-1557. [PMID: 27583823 DOI: 10.1111/jam.13286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 11/28/2022]
Abstract
AIM To purify and characterize antimicrobial peptides derived from the acid extract of Gallus gallus blood cells. METHODS AND RESULTS Two polypeptides (i.e. CHb-1 and CHb-2) with antibacterial activity were detected in the acidic extract of blood cells from chicken (G. gallus). The isolated peptides that possessed a potent antibacterial activity were purified using a two-step chromatography procedure that involved solid-phase extraction of a total protein/peptide extract followed by thin fractionation by reversed-phase high performance liquid chromatography (RP-HPLC). The molecular masses of the purified peptides were similar and were 4824·4 and 4825·2 Da, which have been measured by matrix-assisted laser desorption/ionization mass spectrometry (MALDI TOF MS). Their amino acid sequences were determined by Edman degradation and showed that the peptides were fully identical to the two fragments of G. gallus α-haemoglobin localized into different subunits (A and D respectively). The peptides were active in micromolar concentrations against Gram-negative Escherichia coli K12 TG1. Using the 1-N-phenylnaphthylamine, the FITC-dextran labelled probes and the live/dead staining allowed to show the hemocidin mode of action and estimate the pore size. CONCLUSION In this study, for the first time, α-haemoglobin from chicken (G. gallus) has been investigated as a donor of the two high homologous native peptide fragments that possess potent antibacterial activity in vitro. These are membrane-active peptides and their mechanism of action against E. coli involves a toroidal pore formation. SIGNIFICANCE AND IMPACT OF THE STUDY The obtained results expand the perception of the role of haemoglobin in a living system, describing it as a source of multifunction substances. Additionally, the data presented in this paper may contribute to the development of new, cost-effective, antimicrobial agents.
Collapse
Affiliation(s)
- A S Vasilchenko
- Institute of Cellular and Intracellular Symbiosis, RAS, Orenburg, Russia
| | - E A Rogozhin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Gause Institute of New Antibiotics, Moscow, Russia
| | | | - O L Kartashova
- Institute of Cellular and Intracellular Symbiosis, RAS, Orenburg, Russia
| | - M V Sycheva
- Institute of Cellular and Intracellular Symbiosis, RAS, Orenburg, Russia.,Orenburg State Agrarian University, Orenburg, Russia
| |
Collapse
|
24
|
Malik E, Dennison SR, Harris F, Phoenix DA. pH Dependent Antimicrobial Peptides and Proteins, Their Mechanisms of Action and Potential as Therapeutic Agents. Pharmaceuticals (Basel) 2016; 9:ph9040067. [PMID: 27809281 PMCID: PMC5198042 DOI: 10.3390/ph9040067] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) are potent antibiotics of the innate immune system that have been extensively investigated as a potential solution to the global problem of infectious diseases caused by pathogenic microbes. A group of AMPs that are increasingly being reported are those that utilise pH dependent antimicrobial mechanisms, and here we review research into this area. This review shows that these antimicrobial molecules are produced by a diverse spectrum of creatures, including vertebrates and invertebrates, and are primarily cationic, although a number of anionic examples are known. Some of these molecules exhibit high pH optima for their antimicrobial activity but in most cases, these AMPs show activity against microbes that present low pH optima, which reflects the acidic pH generally found at their sites of action, particularly the skin. The modes of action used by these molecules are based on a number of major structure/function relationships, which include metal ion binding, changes to net charge and conformational plasticity, and primarily involve the protonation of histidine, aspartic acid and glutamic acid residues at low pH. The pH dependent activity of pore forming antimicrobial proteins involves mechanisms that generally differ fundamentally to those used by pH dependent AMPs, which can be described by the carpet, toroidal pore and barrel-stave pore models of membrane interaction. A number of pH dependent AMPs and antimicrobial proteins have been developed for medical purposes and have successfully completed clinical trials, including kappacins, LL-37, histatins and lactoferrin, along with a number of their derivatives. Major examples of the therapeutic application of these antimicrobial molecules include wound healing as well as the treatment of multiple cancers and infections due to viruses, bacteria and fungi. In general, these applications involve topical administration, such as the use of mouth washes, cream formulations and hydrogel delivery systems. Nonetheless, many pH dependent AMPs and antimicrobial proteins have yet to be fully characterized and these molecules, as a whole, represent an untapped source of novel biologically active agents that could aid fulfillment of the urgent need for alternatives to conventional antibiotics, helping to avert a return to the pre-antibiotic era.
Collapse
Affiliation(s)
- Erum Malik
- School of Forensic and Applied Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - Sarah R Dennison
- School of Pharmacy and Biological Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - Frederick Harris
- School of Forensic and Applied Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| | - David A Phoenix
- Office of the Vice Chancellor, London South Bank University, 103 Borough Road, London SE1 0AA, UK.
| |
Collapse
|
25
|
Martinez OF, Agbale CM, Nomiyama F, Franco OL. Deciphering bioactive peptides and their action mechanisms through proteomics. Expert Rev Proteomics 2016; 13:1007-1016. [PMID: 27650042 DOI: 10.1080/14789450.2016.1238305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Bioactive peptides such as antimicrobial peptides (AMPs), ribosomally synthesized and post translationally modified peptides (RiPPs) and the non-ribosomal peptides (NRPs) have emerged with promising applications in medicine, agriculture and industry. However, their development has been limited by several difficulties making it necessary to search for novel discovery methods. In this context, proteomics has been considered a reliable tool. Areas covered: This review highlights recent developments in proteomic tools that facilitate the discovery of AMPs, RiPPs and NRPs as well as the elucidation of action mechanisms of AMPs and resistance mechanisms of pathogens to them. Expert commentary: Proteomic approaches have emerged as useful tools for the study of bioactive peptides, especially mass spectrometry-based peptidomics profiling, a promising strategy for AMP discovery. Furthermore, the rapidly expanding fields of genome mining and genome sequencing techniques, as well as mass spectrometry, have revolutionized the discovery of novel RiPPs and NRPs from complex biological samples.
Collapse
Affiliation(s)
- Osmel Fleitas Martinez
- a Pos-Graduação em Patologia olecular , Universidade de Brasilia , Brasilia-DF Brazil.,b Centro de Analises Proteomicas e Bioquimicas, Programa de Pos-Graduacao em Ciencias Genomicas e Biotecnologia , Universidade Catolica de Brasilia , Brasília , Brazil
| | - Caleb Mawuli Agbale
- c S-Inova Biotech, Programa de Pos-Graduacao em Biotecnologia , Universidade Catolica Dom Bosco , Campo Grande , Brazil.,d Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences , University of Cape Coast , Cape Coast , Ghana
| | - Fernanda Nomiyama
- b Centro de Analises Proteomicas e Bioquimicas, Programa de Pos-Graduacao em Ciencias Genomicas e Biotecnologia , Universidade Catolica de Brasilia , Brasília , Brazil
| | - Octávio Luiz Franco
- a Pos-Graduação em Patologia olecular , Universidade de Brasilia , Brasilia-DF Brazil.,b Centro de Analises Proteomicas e Bioquimicas, Programa de Pos-Graduacao em Ciencias Genomicas e Biotecnologia , Universidade Catolica de Brasilia , Brasília , Brazil.,c S-Inova Biotech, Programa de Pos-Graduacao em Biotecnologia , Universidade Catolica Dom Bosco , Campo Grande , Brazil
| |
Collapse
|