1
|
Peled A, Sprecher E. Proteolytic and Antiproteolytic Activity in the Skin: Gluing the Pieces Together. J Invest Dermatol 2024; 144:466-473. [PMID: 37865898 DOI: 10.1016/j.jid.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 10/23/2023]
Abstract
Epidermal differentiation is ultimately aimed at the formation of a functional barrier capable of protecting the organism from the environment while preventing loss of biologically vital elements. Epidermal differentiation entails a delicately regulated process of cell-cell junction formation and dissolution to enable upward cell migration and desquamation. Over the past two decades, the deciphering of the genetic basis of a number of inherited conditions has delineated the pivotal role played in this process by a series of proteases and protease inhibitors, including serpins, cathepsins, and cystatins, suggesting novel avenues for therapeutic intervention in both rare and common disorders of cornification.
Collapse
Affiliation(s)
- Alon Peled
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
2
|
Lourenço EMG, da Silva F, das Neves AR, Bonfá IS, Ferreira AMT, Menezes ACG, da Silva MEC, Dos Santos JT, Martines MAU, Perdomo RT, Toffoli-Kadri MC, G Barbosa E, Saba S, Beatriz A, Rafique J, de Arruda CCP, de Lima DP. Investigation of the Potential Targets behind the Promising and Highly Selective Antileishmanial Action of Synthetic Flavonoid Derivatives. ACS Infect Dis 2023; 9:2048-2061. [PMID: 37772925 DOI: 10.1021/acsinfecdis.3c00336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Leishmaniases are among the neglected tropical diseases that still cause devastating health, social, and economic consequences to more than 350 million people worldwide. Despite efforts to combat these vector-borne diseases, their incidence does not decrease. Meanwhile, current antileishmanial drugs are old and highly toxic, and safer presentations are unaffordable to the most severely affected human populations. In a previous study by our research group, we synthesized 17 flavonoid derivatives that demonstrated impressive inhibition capacity against rCPB2.8, rCPB3, and rH84Y. These cysteine proteases are highly expressed in the amastigote stage, the target form of the parasite. However, although these compounds have been already described in the literature, until now, the amastigote effect of any of these molecules has not been proven. In this work, we aimed to deeply analyze the antileishmanial action of this set of synthetic flavonoid derivatives by correlating their ability to inhibit cysteine proteases with the action against the parasite. Among all the synthesized flavonoid derivatives, 11 of them showed high activity against amastigotes of Leishmania amazonensis, also providing safety to mammalian host cells. Furthermore, the high production of nitric oxide by infected cells treated with the most active cysteine protease B (CPB) inhibitors confirms a potential immunomodulatory response of macrophages. Besides, considering flavonoids as multitarget drugs, we also investigated other potential antileishmanial mechanisms. The most active compounds were selected to investigate another potential biological pathway behind their antileishmanial action using flow cytometry analysis. The results confirmed an oxidative stress after 48 h of treatment. These data represent an important step toward the validation of CPB as an antileishmanial target, as well as aiding in new drug discovery studies based on this protease.
Collapse
Affiliation(s)
- Estela M G Lourenço
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Fernanda da Silva
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Amarith R das Neves
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Iluska S Bonfá
- Laboratório de Farmacologia e Inflamação, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79074-460 MS, Brazil
| | - Alda Maria T Ferreira
- Laboratório de Imunologia, Biologia Molecular e Bioensaios Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Adriana C G Menezes
- Biotério Central, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Maria E C da Silva
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Jéssica T Dos Santos
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Marco A U Martines
- Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Renata T Perdomo
- Laboratório de Biologia Molecular e Cultura de Células, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Mônica C Toffoli-Kadri
- Laboratório de Farmacologia e Inflamação, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79074-460 MS, Brazil
| | - Euzébio G Barbosa
- Laboratório de Química Farmacêutica Computacional, Departamento de Farmácia, Universidade Federal do Rio Grande do Norte, Natal, 59012-570, RN, Brazil
| | - Sumbal Saba
- Laboratório de Síntese Sustentável e Organocalcogênio - LabSO, Instituto de Química, Universidade Federal de Goiás-UFG, Goiânia, 74690-900 GO, Brazil
| | - Adilson Beatriz
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Jamal Rafique
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
- Laboratório de Síntese Sustentável e Organocalcogênio - LabSO, Instituto de Química, Universidade Federal de Goiás-UFG, Goiânia, 74690-900 GO, Brazil
| | - Carla C P de Arruda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Dênis P de Lima
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| |
Collapse
|
3
|
Malovitski K, Sarig O, Feller Y, Bergson S, Assaf S, Mohamad J, Pavlovsky M, Giladi M, Sprecher E. Defective cathepsin Z affects EGFR expression and causes autosomal dominant palmoplantar keratoderma. Br J Dermatol 2023; 189:302-311. [PMID: 37210216 DOI: 10.1093/bjd/ljad167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND The abnormal function of epidermal growth factor receptor (EGFR) has recently been shown to underlie various disorders of cornification. OBJECTIVES To delineate the genetic basis of a novel dominant form of palmoplantar keratoderma (PPK). METHODS Whole-exome (WES) and direct sequencing, quantitative real-time polymerase chain reaction, protein modelling, confocal immunofluorescence microscopy, immunoblotting, three-dimensional skin equivalents and an enzyme activity assay were used to delineate the genetic basis of a novel dominant form of PPK. RESULTS WES revealed heterozygous variants (c.274T > C and c.305C > T) in CTSZ (encoding cathepsin Z) in four individuals (belonging to three unrelated families) with focal PPK. Bioinformatics and protein modelling predicted the variants to be pathogenic. Previous studies have suggested that EGFR expression may be subject to cathepsin regulation. Immunofluorescence revealed reduced cathepsin Z expression in the upper epidermal layers and concomitant increased epidermal EGFR expression in patients harbouring CTSZ variants. Accordingly, human keratinocytes transfected with constructs expressing PPK-causing variants in CTSZ displayed reduced cathepsin Z enzymatic activity, as well as increased EGFR expression. In line with the role played by EGFR in the regulation of keratinocyte proliferation, human keratinocytes transfected with the PPK-causing variants showed significantly increased proliferation that was abolished upon exposure to erlotinib, an EGFR inhibitor. Similarly, downregulation of CTSZ resulted in increased EGFR expression and increased proliferation in human keratinocytes, suggestive of a loss-of-function effect of the pathogenic variants. Finally, three-dimensional organotypic skin equivalents grown from CTSZ-downregulated cells showed increased epidermal thickness and EGFR expression as seen in patient skin; here, too, erlotinib was found to rescue the abnormal phenotype. CONCLUSIONS Taken collectively, these observations attribute to cathepsin Z a hitherto unrecognized function in epidermal differentiation.
Collapse
Affiliation(s)
- Kiril Malovitski
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Yarden Feller
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shir Bergson
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sari Assaf
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Janan Mohamad
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Moshe Giladi
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Internal Medicine D, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Kumari D, Mahajan S, Kour P, Singh K. Virulence factors of Leishmania parasite: Their paramount importance in unraveling novel vaccine candidates and therapeutic targets. Life Sci 2022; 306:120829. [PMID: 35872004 DOI: 10.1016/j.lfs.2022.120829] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a neglected tropical disease and remains a global concern for healthcare. It is caused by an opportunistic protozoan parasite belonging to the genus Leishmania and affects millions worldwide. This disease is mainly prevalent in tropical and subtropical regions and is associated with a high risk of public morbidity and mortality if left untreated. Transmission of this deadly disease is aggravated by the bite of female sand-fly vectors (Phlebotomus and Lutzomyia). With time, significant advancement in leishmaniasis-related research has been carried out to cope with the disease burden. Still, the Leishmania parasite has also co-evolved with its host and adapted successfully within the host's lethal milieu/environment. Thus, understanding and knowledge of various leishmanial virulence factors responsible for the parasitic infection are essential for exploring drug targets and vaccine candidates. The present review elucidates the importance of virulence factors in pathogenesis and summarizes the major leishmanial virulence molecules contributing to the parasitic infection during host-pathogen interaction. Furthermore, we have also elaborated on the potential contribution of leishmanial virulence proteins in developing vaccine candidates and exploring novel therapeutics against this parasitic disease. We aim to represent a clearer picture of parasite pathogenesis within the human host that can further aid in unraveling new strategies to fight against the deadly infection of leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shavi Mahajan
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Parampreet Kour
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Fromme JE, Zigrino P. The Role of Extracellular Matrix Remodeling in Skin Tumor Progression and Therapeutic Resistance. Front Mol Biosci 2022; 9:864302. [PMID: 35558554 PMCID: PMC9086898 DOI: 10.3389/fmolb.2022.864302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix remodeling in the skin results from a delicate balance of synthesis and degradation of matrix components, ensuring tissue homeostasis. These processes are altered during tumor invasion and growth, generating a microenvironment that supports growth, invasion, and metastasis. Apart from the cellular component, the tumor microenvironment is rich in extracellular matrix components and bound factors that provide structure and signals to the tumor and stromal cells. The continuous remodeling in the tissue compartment sustains the developing tumor during the various phases providing matrices and proteolytic enzymes. These are produced by cancer cells and stromal fibroblasts. In addition to fostering tumor growth, the expression of specific extracellular matrix proteins and proteinases supports tumor invasion after the initial therapeutic response. Lately, the expression and structural modification of matrices were also associated with therapeutic resistance. This review will focus on the significant alterations in the extracellular matrix components and the function of metalloproteinases that influence skin cancer progression and support the acquisition of therapeutic resistance.
Collapse
Affiliation(s)
- Julia E. Fromme
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- *Correspondence: Paola Zigrino,
| |
Collapse
|
6
|
Naeem M, Manzoor S, Abid MUH, Tareen MBK, Asad M, Mushtaq S, Ehsan N, Amna D, Xu B, Hazafa A. Fungal Proteases as Emerging Biocatalysts to Meet the Current Challenges and Recent Developments in Biomedical Therapies: An Updated Review. J Fungi (Basel) 2022; 8:109. [PMID: 35205863 PMCID: PMC8875690 DOI: 10.3390/jof8020109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
With the increasing world population, demand for industrialization has also increased to fulfill humans' living standards. Fungi are considered a source of essential constituents to produce the biocatalytic enzymes, including amylases, proteases, lipases, and cellulases that contain broad-spectrum industrial and emerging applications. The present review discussed the origin, nature, mechanism of action, emerging aspects of genetic engineering for designing novel proteases, genome editing of fungal strains through CRISPR technology, present challenges and future recommendations of fungal proteases. The emerging evidence revealed that fungal proteases show a protective role to many environmental exposures and discovered that an imbalance of protease inhibitors and proteases in the epithelial barriers leads to the protection of chronic eosinophilic airway inflammation. Moreover, mitoproteases recently were found to execute intense proteolytic processes that are crucial for mitochondrial integrity and homeostasis function, including mitochondrial biogenesis, protein synthesis, and apoptosis. The emerging evidence revealed that CRISPR/Cas9 technology had been successfully developed in various filamentous fungi and higher fungi for editing of specific genes. In addition to medical importance, fungal proteases are extensively used in different industries such as foods to prepare butter, fruits, juices, and cheese, and to increase their shelf life. It is concluded that hydrolysis of proteins in industries is one of the most significant applications of fungal enzymes that led to massive usage of proteomics.
Collapse
Affiliation(s)
- Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang 050025, China;
| | - Saba Manzoor
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan;
| | | | | | - Mirza Asad
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| | - Sajida Mushtaq
- Department of Zoology, Government College Women University, Sialkot 51040, Pakistan;
| | - Nazia Ehsan
- Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| | - Dua Amna
- Institute of Food Science & Nutrition, Bahauddin Zakariya University, Multan 60800, Pakistan;
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University (BNU-HKBU) United International College, Zhuhai 519087, China
| | - Abu Hazafa
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| |
Collapse
|
7
|
Ding W, Fan L, Tian Y, He C. Study of the protective effects of cosmetic ingredients on the skin barrier, based on the expression of barrier-related genes and cytokines. Mol Biol Rep 2021; 49:989-995. [PMID: 34799820 PMCID: PMC8825566 DOI: 10.1007/s11033-021-06918-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/30/2021] [Indexed: 11/05/2022]
Abstract
Background Sensitive skin is the result of a complex process that is closely linked to the damage of the skin barrier. There are no recognized methods for evaluating the efficacy of anti-allergy products. Methods In this study, a model of skin barrier damage was created by treating HaCaT cells with 60 μg/ml of sodium dodecyl sulfate for 48 h. The protective effects of nine cosmetic ingredients, including oat extract (S1), on the skin barrier were investigated based on the gene expression levels of aquaporin3 (AQP3), filaggrin (FLG), caspase-14 (CASP14), and human tissue kallikrein7 (KLK7), as well as those of various interleukins (IL) and vascular endothelial growth factor (VEGF). Results Among the nine ingredients, S1 had a good protective effect on the function of the skin barrier. It promoted the expression of AQP3, FLG, and CASP14, while inhibiting the expression of KLK7 in HaCaT cells, at a concentration of 0.06%. It also maintained IL-6, IL-8, and VEGF at appropriate levels while promoting the proliferation and differentiation of HaCaT cells. Conclusions The above indicators allow for the preliminary establishment of a method to evaluate the efficacy of the barrier protection ability of sensitive skin. Supplementary Information The online version contains supplementary material available at 10.1007/s11033-021-06918-5.
Collapse
Affiliation(s)
- Wenyu Ding
- Cosmetics Department, College of Chemistry and Materials Engineering, Beijing Technology and Business University, No. 11 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Linna Fan
- Cosmetics Department, College of Chemistry and Materials Engineering, Beijing Technology and Business University, No. 11 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Yan Tian
- Air Force Medical Center, PLA, Beijing, 100142, China
| | - Congfen He
- Cosmetics Department, College of Chemistry and Materials Engineering, Beijing Technology and Business University, No. 11 Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
8
|
Xu TT, Zeng XW, Wang XH, Yang LX, Luo G, Yu T. Cystatin-B Negatively Regulates the Malignant Characteristics of Oral Squamous Cell Carcinoma Possibly Via the Epithelium Proliferation/Differentiation Program. Front Oncol 2021; 11:707066. [PMID: 34504787 PMCID: PMC8421684 DOI: 10.3389/fonc.2021.707066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Disturbance in the proteolytic process is one of the malignant signs of tumors. Proteolysis is highly orchestrated by cysteine cathepsin and its inhibitors. Cystatin-B (CSTB) is a general cysteine cathepsin inhibitor that prevents cysteine cathepsin from leaking from lysosomes and causing inappropriate proteolysis. Our study found that CSTB was downregulated in both oral squamous cell carcinoma (OSCC) tissues and cells compared with normal controls. Immunohistochemical analysis showed that CSTB was mainly distributed in the epithelial structure of OSCC tissues, and its expression intensity was related to the grade classification. A correlation analysis between CSTB and clinical prognosis was performed using gene expression data and clinical information acquired from The Cancer Genome Atlas (TCGA) database. Patients with lower expression levels of CSTB had shorter disease-free survival times and poorer clinicopathological features (e.g., lymph node metastases, perineural invasion, low degree of differentiation, and advanced tumor stage). OSCC cell models overexpressing CSTB were constructed to assess the effects of CSTB on malignant biological behaviors and upregulation of CSTB inhibited cell proliferation, migration, and invasion in vitro. Weighted gene correlation network analysis (WGCNA) and gene set enrichment analysis (GSEA) were performed based on the TCGA data to explore potential mechanisms, and CSTB appeared to correlate with squamous epithelial proliferation-differentiation processes, such as epidermal cell differentiation and keratinization. Moreover, in WGCNA, the gene module most associated with CSTB expression (i.e., the brown module) was also the one most associated with grade classification. Upregulation of CSTB promoted the expression levels of markers (LOR, IVL, KRT5/14, and KRT1/10), reflecting a tendency for differentiation and keratinization in vitro. Gene expression profile data of the overexpressed CSTB cell line were obtained by RNA sequencing (RNA-seq) technology. By comparing the GSEA enrichment results of RNA-seq data (from the OSCC models overexpressing CSTB) and existing public database data, three gene sets (i.e., apical junction, G2/M checkpoint, etc.) and six pathways (e.g., NOTCH signaling pathway, glycosaminoglycan degradation, mismatch repair, etc.) were enriched in the data from both sources. Overall, our study shows that CSTB is downregulated in OSCC and might regulate the malignant characteristics of OSCC via the epithelial proliferation/differentiation program.
Collapse
Affiliation(s)
- Tian-Tian Xu
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xiao-Wen Zeng
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xin-Hong Wang
- Department of Oral Pathology and Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lu-Xi Yang
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Gang Luo
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Ting Yu
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
9
|
Vidal-Quist JC, Ortego F, Hernández-Crespo P. Contribution of cysteine and serine proteases to proteolytic digestion in an allergy-eliciting house dust mite. JOURNAL OF INSECT PHYSIOLOGY 2021; 133:104285. [PMID: 34284041 DOI: 10.1016/j.jinsphys.2021.104285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 06/13/2023]
Abstract
The digestive physiology of house dust mites (HDM) is of interest to understand their allergenicity towards humans since many of their allergens are digestive enzymes and/or are excreted into airborne fecal pellets. The aim of this study is to provide insight on the biochemical basis of proteolytic digestion in Dermatophagoides pteronyssinus, the most widespread HDM species. First, assays using non-specific protein substrates on purified fecal and body extracts determined that body-associated activity is almost exclusively dependent on cysteine proteases, and specifically on major allergen Der p 1. By contrast, cysteine and serine proteases contributed similarly to the activity estimated on fecal extracts. Second, the screening of group-specific peptide-based protease inhibitors followed by ingestion bioassays revealed that the human skin-derived cysteine protease inhibitor cystatin A produces a significant reduction in mite feeding (i.e. excreted guanine), and triggers the overproduction of Der p 1 (3-fold increase by ELISA). Noteworthy, the inhibition of cysteine proteases by cystatin A also resulted in a reduction in three non-target serine protease activities. Further incubation of these extracts with exogenous Der p 1, but not with other commercial cysteine proteases, restored trypsin (Der p 3) and chymotrypsin (Der p 6) activities, indicating that Der p 1 is responsible for their activation in vivo. Finally, the role of serine proteases on the mite's digestive physiology is discussed based on their remarkable activity in fecal extracts and the autocoprophagic behavior reported in mites in this study.
Collapse
Affiliation(s)
- José Cristian Vidal-Quist
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain.
| | - Félix Ortego
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain
| | - Pedro Hernández-Crespo
- Laboratorio de Entomología Aplicada a la Agricultura y la Salud, Centro de Investigaciones Biológicas Margarita Salas CSIC, Spain
| |
Collapse
|
10
|
Eckl KM, Gruber R, Brennan L, Marriott A, Plank R, Moosbrugger-Martinz V, Blunder S, Schossig A, Altmüller J, Thiele H, Nürnberg P, Zschocke J, Hennies HC, Schmuth M. Cystatin M/E Variant Causes Autosomal Dominant Keratosis Follicularis Spinulosa Decalvans by Dysregulating Cathepsins L and V. Front Genet 2021; 12:689940. [PMID: 34322157 PMCID: PMC8312243 DOI: 10.3389/fgene.2021.689940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
Keratosis follicularis spinulosa decalvans (KFSD) is a rare cornification disorder with an X-linked recessive inheritance in most cases. Pathogenic variants causing X-linked KFSD have been described in MBTPS2, the gene for a membrane-bound zinc metalloprotease that is involved in the cleavage of sterol regulatory element binding proteins important for the control of transcription. Few families have been identified with an autosomal dominant inheritance of KFSD. We present two members of an Austrian family with a phenotype of KFSD, a mother and her son. The disease was not observed in her parents, pointing to a dominant inheritance with a de novo mutation in the index patient. Using whole-exome sequencing, we identified a heterozygous missense variant in CST6 in DNA samples from the index patient and her affected son. In line with family history, the variant was not present in samples from her parents. CST6 codes for cystatin M/E, a cysteine protease inhibitor. Patient keratinocytes showed increased expression of cathepsin genes CTSL and CTSV and reduced expression of transglutaminase genes TGM1 and TGM3. A relative gain of active, cleaved transglutaminases was found in patient keratinocytes compared to control cells. The variant found in CST6 is expected to affect protein targeting and results in marked disruption of the balance between cystatin M/E activity and its target proteases and eventually transglutaminases 1 and 3. This disturbance leads to an impairment of terminal epidermal differentiation and proper hair shaft formation seen in KFSD.
Collapse
Affiliation(s)
- Katja M. Eckl
- Department of Biology, Edge Hill University, Ormskirk, United Kingdom
| | - Robert Gruber
- Department of Dermatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Louise Brennan
- Department of Biology, Edge Hill University, Ormskirk, United Kingdom
| | - Andrew Marriott
- Department of Biology, Edge Hill University, Ormskirk, United Kingdom
| | - Roswitha Plank
- Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Stefan Blunder
- Department of Dermatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Schossig
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Altmüller
- Cologne Center for Genomics, Faculty of Medicine and Cologne University Hospital, University of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics, Faculty of Medicine and Cologne University Hospital, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, Faculty of Medicine and Cologne University Hospital, University of Cologne, Cologne, Germany
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hans Christian Hennies
- Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Cologne Center for Genomics, Faculty of Medicine and Cologne University Hospital, University of Cologne, Cologne, Germany
| | - Matthias Schmuth
- Department of Dermatology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Cystatin M/E (Cystatin 6): A Janus-Faced Cysteine Protease Inhibitor with Both Tumor-Suppressing and Tumor-Promoting Functions. Cancers (Basel) 2021; 13:cancers13081877. [PMID: 33919854 PMCID: PMC8070812 DOI: 10.3390/cancers13081877] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Alongside its contribution in maintaining skin homeostasis and its probable involvement in fetal and placental development, cystatin M/E (also known as cystatin 6) was first described as a tumor suppressor of breast cancer. This review aims to provide an update on cystatin M/E with particular attention paid to its role during tumorigenesis. Cystatin M/E, which is related to type 2 cystatins, displays the unique property of being a dual tight-binding inhibitor of both legumain (also known as asparagine endopeptidase) and cysteine cathepsins L, V and B, while its expression level is epigenetically regulated via the methylation of the CST6 promoter region. The tumor-suppressing role of cystatin M/E was further reported in melanoma, cervical, brain, prostate, gastric and renal cancers, and cystatin M/E was proposed as a biomarker of prognostic significance. Contrariwise, cystatin M/E could have an antagonistic function, acting as a tumor promoter (e.g., oral, pancreatic cancer, thyroid and hepatocellular carcinoma). Taking into account these apparently divergent functions, there is an urgent need to decipher the molecular and cellular regulatory mechanisms of the expression and activity of cystatin M/E associated with the safeguarding homeostasis of the proteolytic balance as well as its imbalance in cancer.
Collapse
|
12
|
Ribeiro JFR, Cianni L, Li C, Warwick TG, de Vita D, Rosini F, Dos Reis Rocho F, Martins FCP, Kenny PW, Lameira J, Leitão A, Emsley J, Montanari CA. Crystal structure of Leishmania mexicana cysteine protease B in complex with a high-affinity azadipeptide nitrile inhibitor. Bioorg Med Chem 2020; 28:115743. [PMID: 33038787 DOI: 10.1016/j.bmc.2020.115743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 11/19/2022]
Abstract
Leishmania mexicana is an obligate intracellular protozoan parasite that causes the cutaneous form of leishmaniasis affecting South America and Mexico. The cysteine protease LmCPB is essential for the virulence of the parasite and therefore, it is an appealing target for antiparasitic therapy. A library of nitrile-based cysteine protease inhibitors was screened against LmCPB to develop a treatment of cutaneous leishmaniasis. Several compounds are sufficiently high-affinity LmCPB inhibitors to serve both as starting points for drug discovery projects and as probes for target validation. A 1.4 Å X ray crystal structure, the first to be reported for LmCPB, was determined for the complex of this enzyme covalently bound to an azadipeptide nitrile ligand. Mapping the structure-activity relationships for LmCPB inhibition revealed superadditive effects for two pairs of structural transformations. Therefore, this work advances our understanding of azadipeptidyl and dipeptidyl nitrile structure-activity relationships for LmCPB structure-based inhibitor design. We also tested the same series of inhibitors on related cysteine proteases cathepsin L and Trypanosoma cruzi cruzain. The modulation of these mammalian and protozoan proteases represents a new framework for targeting papain-like cysteine proteases.
Collapse
Affiliation(s)
- Jean F R Ribeiro
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Lorenzo Cianni
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Chan Li
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Thomas G Warwick
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Daniela de Vita
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Fabiana Rosini
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Fernanda Dos Reis Rocho
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Felipe C P Martins
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Peter W Kenny
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Jeronimo Lameira
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil; Laboratory of Design and Development of Pharmaceuticals, Federal University of Pará, Belém, Brazil
| | - Andrei Leitão
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Jonas Emsley
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK.
| | - Carlos A Montanari
- Medicinal and Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, Brazil.
| |
Collapse
|
13
|
Whole-genome Sequencing and Mining of Protease Coding Genes in Bacillus paralicheniformis MKU3, and its Degradomics in Feather Meal Medium. Curr Microbiol 2020; 78:206-217. [PMID: 33175193 DOI: 10.1007/s00284-020-02271-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/22/2020] [Indexed: 10/23/2022]
Abstract
Bacillus paralicheniformis MKU3 produces commercially important keratinolytic proteases by utilizing chicken feather. To unravel the genetics of these degrading keratinolytic proteases in B. paralicheniformis MKU3, we sequenced the genome of this bacterium and studied the protease distribution and their characteristics using bioinformatics tools. Also, a proteomic analysis was performed to identify the consortium of proteases involved in feather hydrolysis. A total of 2,531,755 quality reads were obtained in whole genome sequencing with an approximate coverage fold of 105. The draft genome consists of 4,370,039 bp with 45 contigs. The draft genome codes for 4874 protein-coding genes. Furthermore, 109 genes coding for RNA, including 26 rRNA and 83 tRNA, were identified. Phylogenetic analysis of B. paralicheniformis MKU3 showed closest homolog to B. paralicheniformis F47. Genes coding for proteases belonging to five families were identified with the following proportions 37%, 36%, 9%, 14%, 2%, and 2% of serine-, metallo-, cysteine-, mixed-, and uncharacterized proteases, respectively. Metallo- and serine-protease represented more than 70% of the total proteases. Major protease families distributed in the genome were S8, S9, S33, M20, M50, C26, and C40. Most of the proteases showed significant similarity with the conserved domain database and also identified conserved catalytic sites and domains. SDS-PAGE and zymogram analysis of concentrated feather hydrolysis revealed the active proteases ranging from 10 to 250 kD in size. Proteomic analysis on the feather hydrolysis of B. paralicheniformis MKU3 identified two proteases belonging to serine proteases (S8) and other two as metalloproteases.
Collapse
|
14
|
Cianni L, Feldmann CW, Gilberg E, Gütschow M, Juliano L, Leitão A, Bajorath J, Montanari CA. Can Cysteine Protease Cross-Class Inhibitors Achieve Selectivity? J Med Chem 2019; 62:10497-10525. [DOI: 10.1021/acs.jmedchem.9b00683] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lorenzo Cianni
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Christian Wolfgang Feldmann
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Erik Gilberg
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Luiz Juliano
- A. C. Camargo Cancer Center and São Paulo Medical School of Federal University of São Paulo, Rua Professor Antônio Prudente, 211, 01509-010 São Paulo, SP, Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Carlos A. Montanari
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| |
Collapse
|
15
|
Magnadóttir B, Bragason BT, Bricknell IR, Bowden T, Nicholas AP, Hristova M, Guðmundsdóttir S, Dodds AW, Lange S. Peptidylarginine deiminase and deiminated proteins are detected throughout early halibut ontogeny - Complement components C3 and C4 are post-translationally deiminated in halibut (Hippoglossus hippoglossus L.). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:1-19. [PMID: 30395876 DOI: 10.1016/j.dci.2018.10.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 06/08/2023]
Abstract
Post-translational protein deimination is mediated by peptidylarginine deiminases (PADs), which are calcium dependent enzymes conserved throughout phylogeny with physiological and pathophysiological roles. Protein deimination occurs via the conversion of protein arginine into citrulline, leading to structural and functional changes in target proteins. In a continuous series of early halibut development from 37 to 1050° d, PAD, total deiminated proteins and deiminated histone H3 showed variation in temporal and spatial detection in various organs including yolksac, muscle, skin, liver, brain, eye, spinal cord, chondrocytes, heart, intestines, kidney and pancreas throughout early ontogeny. For the first time in any species, deimination of complement components C3 and C4 is shown in halibut serum, indicating a novel mechanism of complement regulation in immune responses and homeostasis. Proteomic analysis of deiminated target proteins in halibut serum further identified complement components C5, C7, C8 C9 and C1 inhibitor, as well as various other immunogenic, metabolic, cytoskeletal and nuclear proteins. Post-translational deimination may facilitate protein moonlighting, an evolutionary conserved phenomenon, allowing one polypeptide chain to carry out various functions to meet functional requirements for diverse roles in immune defences and tissue remodelling.
Collapse
Affiliation(s)
- Bergljót Magnadóttir
- Institute for Experimental Pathology, University of Iceland, Keldur v. Vesturlandsveg, 112 Reykjavik, Iceland.
| | - Birkir Thor Bragason
- Institute for Experimental Pathology, University of Iceland, Keldur v. Vesturlandsveg, 112 Reykjavik, Iceland.
| | - Ian R Bricknell
- Aquaculture Research Institute School of Marine Sciences, University of Maine, Orono, ME, USA.
| | - Timothy Bowden
- Aquaculture Research Institute School of Food & Agriculture, University of Maine, University of Maine, Orono, ME, USA.
| | - Anthony P Nicholas
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Mariya Hristova
- Perinatal Brain Protection and Repair Group, EGA Institute for Women's Health, University College London, London, WC1E 6HX, UK.
| | - Sigríður Guðmundsdóttir
- Institute for Experimental Pathology, University of Iceland, Keldur v. Vesturlandsveg, 112 Reykjavik, Iceland.
| | - Alister W Dodds
- MRC Immunochemistry Unit, Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London, W1W 6UW, UK.
| |
Collapse
|
16
|
Tacheau C, Weisgerber F, Fagot D, Bastien P, Verdier MP, Liboutet M, Sore G, Bernard BA. Vichy Thermal Spring Water (VTSW), a cosmetic ingredient of potential interest in the frame of skin ageing exposome: anin vitrostudy. Int J Cosmet Sci 2018; 40:377-387. [DOI: 10.1111/ics.12470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/31/2018] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - D. Fagot
- L'Oréal R&I; Aulnay-sous-Bois; France
| | | | | | | | | | | |
Collapse
|
17
|
Föll MC, Fahrner M, Gretzmeier C, Thoma K, Biniossek ML, Kiritsi D, Meiss F, Schilling O, Nyström A, Kern JS. Identification of tissue damage, extracellular matrix remodeling and bacterial challenge as common mechanisms associated with high-risk cutaneous squamous cell carcinomas. Matrix Biol 2017; 66:1-21. [PMID: 29158163 DOI: 10.1016/j.matbio.2017.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
In this study we used a genetic extracellular matrix (ECM) disease to identify mechanisms associated with aggressive behavior of cutaneous squamous cell carcinoma (cSCC). cSCC is one of the most common malignancies and usually has a good prognosis. However, some cSCCs recur or metastasize and cause significant morbidity and mortality. Known factors that are associated with aggressiveness of cSCCs include tumor grading, size, localization and microinvasive behavior. To investigate molecular mechanisms that influence biologic behavior we used global proteomic and histologic analyses of formalin-fixed paraffin-embedded tissue of primary human cSCCs. We compared three groups: non-recurring, non-metastasizing low-risk sporadic cSCCs; metastasizing sporadic cSCCs; and cSCCs from patients with recessive dystrophic epidermolysis bullosa (RDEB). RDEB is a genetic skin blistering and ECM disease caused by collagen VII deficiency. Patients commonly suffer from high-risk early onset cSCCs that frequently metastasize. The results indicate that different processes are associated with formation of RDEB cSCCs compared to sporadic cSCCs. Sporadic cSCCs show signs of UV damage, whereas RDEB cSCCs have higher mutational rates and display tissue damage, inflammation and subsequent remodeling of the dermal ECM as tumor initiating factors. Interestingly the two high-risk groups - high-risk metastasizing sporadic cSCCs and RDEB cSCCs - are both associated with tissue damage and ECM remodeling in gene-ontology enrichment and Search Tool for the Retrieval of Interacting Genes/Proteins analyses. In situ histologic analyses validate these results. The high-risk cSCCs also show signatures of enhanced bacterial challenge. Histologic analyses confirm correlation of bacterial colonization with worse prognosis. Collectively, this unbiased study - performed directly on human patient material - reveals that common microenvironmental alterations linked to ECM remodeling and increased bacterial challenges are denominators of high-risk cSCCs. The proteins identified here could serve as potential diagnostic markers and therapeutic targets in high-risk cSCCs.
Collapse
Affiliation(s)
- Melanie C Föll
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany
| | - Matthias Fahrner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Germany
| | - Christine Gretzmeier
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Käthe Thoma
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany
| | - Dimitra Kiritsi
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Frank Meiss
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany.
| | - Johannes S Kern
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany; Department of Dermatology, Royal Melbourne Hospital, Parkville and Box Hill Hospital - Monash University, Eastern Health Clinical School, Box Hill, Victoria, Australia
| |
Collapse
|
18
|
Cai D, Li Y, Zhou C, Jiang Y, Jiao J, Wu L. Comparative proteomics analysis of primary cutaneous amyloidosis. Exp Ther Med 2017; 14:3004-3012. [PMID: 28912854 PMCID: PMC5585729 DOI: 10.3892/etm.2017.4852] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 05/25/2017] [Indexed: 01/02/2023] Open
Abstract
Primary cutaneous amyloidosis (PCA) is a localized skin disorder that is characterized by the abnormal deposition of amyloid in the extracellular matrix (ECM) of the dermis. The pathogenesis of PCA is poorly understood. The objective of the present study was to survey proteome changes in PCA lesions in order to gain insight into the molecular basis and pathogenesis of PCA. Total protein from PCA lesions and normal skin tissue samples were extracted and analyzed using the isobaric tags for relative and absolute quantitation technique. The function of differentially expressed proteins in PCA were analyzed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction analysis. The proteins that were most upregulated in PCA lesions were further analyzed by immunohistochemistry. A total of 1,032 proteins were identified in PCA lesions and control skin samples, with 51 proteins differentially expressed in PCA lesions, of which 27 were upregulated. In PCA lesions, the upregulated proteins were primarily extracellulary located. In addition, GO analysis indicated that the upregulated proteins were significantly enriched in the biological processes of epidermal development, collagen fiber organization and response to wounding (adjusted P<0.001). KEGG analysis indicated that the upregulated proteins were significantly enriched in the signaling pathways of cell communication, ECM receptor interaction and focal adhesion (adjusted P<0.001). Furthermore, the upregulated proteins were enriched in the molecular function of calcium ion binding, and the calcium binding proteins calmodulin-like protein 5, S100 calcium-binding protein A7 (S100A7)/fatty-acid binding protein and S100A8/A9 exhibited the highest levels of upregulation in PCA. This analysis of differentially expressed proteins in PCA suggests that increased focal adhesion, differentiation and wound healing is associated with the pathogenesis of PCA.
Collapse
Affiliation(s)
- Daxing Cai
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yang Li
- Beijing Protein Innovation Co. Ltd., Beijing 101318, P.R. China
| | - Chunlei Zhou
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yulin Jiang
- Beijing Protein Innovation Co. Ltd., Beijing 101318, P.R. China
| | - Jian Jiao
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lin Wu
- Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| |
Collapse
|
19
|
Vidal-Quist JC, Ortego F, Rombauts S, Castañera P, Hernández-Crespo P. Dietary shifts have consequences for the repertoire of allergens produced by the European house dust mite. MEDICAL AND VETERINARY ENTOMOLOGY 2017; 31:272-280. [PMID: 28429373 DOI: 10.1111/mve.12234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/12/2017] [Accepted: 01/30/2017] [Indexed: 06/07/2023]
Abstract
Products manufactured from mass-cultured house dust mites, currently commercialized for the diagnosis and immunotherapy of allergy, are heterogeneous in terms of allergen composition and thus present concerns to regulatory authorities. The most abundant species, Dermatophagoides pteronyssinus (Trouessart) (Astigmata: Pyroglyphidae), produces 19 allergenic proteins. Many of these are putatively involved in mite digestive physiology and metabolism. This study aimed to evaluate the effects of mite-rearing media on allergen production. Mites were adapted to feed on culture media supplemented with proteins, lipids, carbohydrates or beard shavings, and collected to quantify major allergens (Der p 1 and 2) by immunodetection, transcription of allergen genes by real-time quantitative polymerase chain reaction, and allergen-related enzymatic activities. All culture media significantly affected the content of major allergens. Modification of macronutrients in the diet produced minor effects on the transcription of allergen genes, but significantly altered mite allergen-related activities. The most remarkable impacts were detected in mites feeding on beard shavings and were reflected in reductions in the content of major allergens, alterations in the transcription of nine allergen genes, and changes in eight allergen-related activities. These results demonstrate the importance of culture media to the quality and consistency of mite extracts used for pharmaceuticals, and highlight the need to further elucidate allergen production by mites in the laboratory and in domestic environments.
Collapse
Affiliation(s)
- J C Vidal-Quist
- Laboratory of Insect-Plant Interaction, Department of Environmental Biology, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - F Ortego
- Laboratory of Insect-Plant Interaction, Department of Environmental Biology, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - S Rombauts
- VIB Center for Plant Systems Biology, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - P Castañera
- Laboratory of Insect-Plant Interaction, Department of Environmental Biology, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - P Hernández-Crespo
- Laboratory of Insect-Plant Interaction, Department of Environmental Biology, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
20
|
Drake MT, Clarke BL, Oursler MJ, Khosla S. Cathepsin K Inhibitors for Osteoporosis: Biology, Potential Clinical Utility, and Lessons Learned. Endocr Rev 2017; 38:325-350. [PMID: 28651365 PMCID: PMC5546879 DOI: 10.1210/er.2015-1114] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 06/20/2017] [Indexed: 12/24/2022]
Abstract
Cathepsin K is a cysteine protease member of the cathepsin lysosomal protease family. Although cathepsin K is highly expressed in osteoclasts, lower levels of cathepsin K are also found in a variety of other tissues. Secretion of cathepsin K from the osteoclast into the sealed osteoclast-bone cell interface results in efficient degradation of type I collagen. The absence of cathepsin K activity in humans results in pycnodysostosis, characterized by increased bone mineral density and fractures. Pharmacologic cathepsin K inhibition leads to continuous increases in bone mineral density for ≤5 years of treatment and improves bone strength at the spine and hip. Compared with other antiresorptive agents, cathepsin K inhibition is nearly equally efficacious for reducing biochemical markers of bone resorption but comparatively less active for reducing bone formation markers. Despite multiple efforts to develop cathepsin K inhibitors, potential concerns related to off-target effects of the inhibitors against other cathepsins and cathepsin K inhibition at nonbone sites, including skin and perhaps cardiovascular and cerebrovascular sites, prolonged the regulatory approval process. A large multinational randomized, double-blind phase III study of odanacatib in postmenopausal women with osteoporosis was recently completed. Although that study demonstrated clinically relevant reductions in fractures at multiple sites, odanacatib was ultimately withdrawn from the regulatory approval process after it was found to be associated with an increased risk of cerebrovascular accidents. Nonetheless, the underlying biology and clinical effects of cathepsin K inhibition remain of considerable interest and could guide future therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Matthew T. Drake
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Bart L. Clarke
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Merry Jo Oursler
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Sundeep Khosla
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|
21
|
Ngcungcu T, Oti M, Sitek JC, Haukanes BI, Linghu B, Bruccoleri R, Stokowy T, Oakeley EJ, Yang F, Zhu J, Sultan M, Schalkwijk J, van Vlijmen-Willems IMJJ, von der Lippe C, Brunner HG, Ersland KM, Grayson W, Buechmann-Moller S, Sundnes O, Nirmala N, Morgan TM, van Bokhoven H, Steen VM, Hull PR, Szustakowski J, Staedtler F, Zhou H, Fiskerstrand T, Ramsay M. Duplicated Enhancer Region Increases Expression of CTSB and Segregates with Keratolytic Winter Erythema in South African and Norwegian Families. Am J Hum Genet 2017; 100:737-750. [PMID: 28457472 DOI: 10.1016/j.ajhg.2017.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 03/27/2017] [Indexed: 12/30/2022] Open
Abstract
Keratolytic winter erythema (KWE) is a rare autosomal-dominant skin disorder characterized by recurrent episodes of palmoplantar erythema and epidermal peeling. KWE was previously mapped to 8p23.1-p22 (KWE critical region) in South African families. Using targeted resequencing of the KWE critical region in five South African families and SNP array and whole-genome sequencing in two Norwegian families, we identified two overlapping tandem duplications of 7.67 kb (South Africans) and 15.93 kb (Norwegians). The duplications segregated with the disease and were located upstream of CTSB, a gene encoding cathepsin B, a cysteine protease involved in keratinocyte homeostasis. Included in the 2.62 kb overlapping region of these duplications is an enhancer element that is active in epidermal keratinocytes. The activity of this enhancer correlated with CTSB expression in normal differentiating keratinocytes and other cell lines, but not with FDFT1 or NEIL2 expression. Gene expression (qPCR) analysis and immunohistochemistry of the palmar epidermis demonstrated significantly increased expression of CTSB, as well as stronger staining of cathepsin B in the stratum granulosum of affected individuals than in that of control individuals. Analysis of higher-order chromatin structure data and RNA polymerase II ChIA-PET data from MCF-7 cells did not suggest remote effects of the enhancer. In conclusion, KWE in South African and Norwegian families is caused by tandem duplications in a non-coding genomic region containing an active enhancer element for CTSB, resulting in upregulation of this gene in affected individuals.
Collapse
Affiliation(s)
- Thandiswa Ngcungcu
- Division of Human Genetics, School of Pathology and the Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Martin Oti
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525 GA, the Netherlands; Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Jan C Sitek
- Department of Dermatology, Oslo University Hospital, Oslo 0424, Norway; Centre for Rare Disorders, Oslo University Hospital, Oslo 0424, Norway
| | - Bjørn I Haukanes
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5021, Norway
| | - Bolan Linghu
- Computational Biomedicine, WRD Genome Sciences & Technologies, Pfizer Worldwide R&D, Cambridge, MA 02139, USA
| | - Robert Bruccoleri
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA; Congenomics, Glastonbury, CT 06033, USA
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, Bergen 5020, Norway
| | - Edward J Oakeley
- Novartis Institutes for BioMedical Research, Basel 4056, Switzerland
| | - Fan Yang
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Jiang Zhu
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Marc Sultan
- Novartis Institutes for BioMedical Research, Basel 4056, Switzerland
| | - Joost Schalkwijk
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Ivonne M J J van Vlijmen-Willems
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | | | - Han G Brunner
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands; Maastricht UMC, Department of Clinical Genetics and School for Oncology and Developmental Biology (GROW), Maastricht 6202 AZ, the Netherlands
| | - Kari M Ersland
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5021, Norway; Department of Clinical Science, University of Bergen, Bergen 5020, Norway
| | - Wayne Grayson
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and Ampath National Laboratories, Johannesburg 2193, South Africa
| | | | - Olav Sundnes
- Department of Dermatology, Oslo University Hospital, Oslo 0424, Norway; Laboratory for Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Nanguneri Nirmala
- Institute for Clinical Research and Policy Studies, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Thomas M Morgan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Hans van Bokhoven
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Vidar M Steen
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5021, Norway; Department of Clinical Science, University of Bergen, Bergen 5020, Norway
| | - Peter R Hull
- Division of Clinical Dermatology and Cutaneous Science, Dalhousie University, Halifax, NS B3H 1V7, Canada
| | | | - Frank Staedtler
- Novartis Institutes for BioMedical Research, Basel 4056, Switzerland
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525 GA, the Netherlands; Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Torunn Fiskerstrand
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5021, Norway; Department of Clinical Science, University of Bergen, Bergen 5020, Norway.
| | - Michele Ramsay
- Division of Human Genetics, School of Pathology and the Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.
| |
Collapse
|
22
|
da Silva RR. Bacterial and Fungal Proteolytic Enzymes: Production, Catalysis and Potential Applications. Appl Biochem Biotechnol 2017; 183:1-19. [DOI: 10.1007/s12010-017-2427-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/24/2017] [Indexed: 11/29/2022]
|
23
|
Lynch AM, Wagner BD, Mandava N, Palestine AG, Mourani PM, McCourt EA, Oliver SCN, Abman SH. The Relationship of Novel Plasma Proteins in the Early Neonatal Period With Retinopathy of Prematurity. Invest Ophthalmol Vis Sci 2016; 57:5076-5082. [PMID: 27679852 PMCID: PMC5053115 DOI: 10.1167/iovs.16-19653] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Retinopathy of prematurity (ROP) is a vision-threatening disease associated with abnormal retinal vascular development. Proteins from the insulin-like growth factor pathway are related to ROP. However, there is a paucity of research on the role of other proteins in ROP. The aim of this study was to identify plasma proteins related to clinically significant ROP. Methods We measured 1121 plasma proteins in the early neonatal period in infants at risk for ROP using an aptamer-based proteomic technology. The primary aim of the study was to compare plasma protein concentrations in infants who did (n = 12) and did not (n = 23) subsequently develop clinically significant ROP using logistic regression. As a secondary aim, we examined patterns in the proteins across categories of clinically significant, low-grade, and no ROP groups. Results Lower levels of 16 proteins were associated with an increased risk of clinically significant ROP. In this group, superoxide dismutase (Mn), mitochondrial (MnSOD), and chordin-like protein 1 (CRDL1) were highly ranked. Other proteins in this group included: C-C motif chemokine 14 (HCC-1), prolactin, insulin-like growth factor-binding protein 7 (IGFBP-7), and eotaxin. Higher levels of 12 proteins were associated with a higher risk for ROP. Fibroblast growth factor 19 (FGF-19) was the top-ranked protein target followed by hepatocyte growth factor-like protein (MSP), luteinizing hormone (LH), cystatin M, plasminogen, and proprotein convertase subtilisin/kexin type 9 (PCSK9). We also noted different patterns in the trend of concentrations of proteins across the clinically significant, low-grade, and no ROP groups. Conclusions We discovered plasma proteins with novel associations with clinically significant ROP (MnSOD, CRDL1, PCSK9), proteins with links to established ROP signaling pathways (IGFBP-7), and proteins such as MnSOD that may be a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Anne M Lynch
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, United States
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Peter M Mourani
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Emily A McCourt
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Scott C N Oliver
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Steven H Abman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
24
|
Fang JY, Wang PW, Huang CH, Chen MH, Wu YR, Pan TL. Skin aging caused by intrinsic or extrinsic processes characterized with functional proteomics. Proteomics 2016; 16:2718-2731. [PMID: 27459910 DOI: 10.1002/pmic.201600141] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/24/2016] [Accepted: 07/21/2016] [Indexed: 12/27/2022]
Abstract
The skin provides protection against environmental stress. However, intrinsic and extrinsic aging causes significant alteration to skin structure and components, which subsequently impairs molecular characteristics and biochemical processes. Here, we have conducted an immunohistological investigation and established the proteome profiles on nude mice skin to verify the specific responses during aging caused by different factors. Our results showed that UVB-elicited aging results in upregulation of proliferating cell nuclear antigen and strong oxidative damage in DNA, whereas chronological aging abolished epidermal cell growth and increased the expression of caspase-14, as well as protein carbonylation. Network analysis indicated that the programmed skin aging activated the ubiquitin system and triggered obvious downregulation of 14-3-3 sigma, which might accelerate the loss of cell growth capacity. On the other hand, UVB stimulation enhanced inflammation and the risk of skin carcinogenesis. Collectively, functional proteomics could provide large-scale investigation of the potent proteins and molecules that play important roles in skin subjected to both intrinsic and extrinsic aging.
Collapse
Affiliation(s)
- Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pei-Wen Wang
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Hsun Huang
- Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yun-Ru Wu
- Graduate Institute of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tai-Long Pan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan. .,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan. .,Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
25
|
Castro-Gomes T, Corrotte M, Tam C, Andrews NW. Plasma Membrane Repair Is Regulated Extracellularly by Proteases Released from Lysosomes. PLoS One 2016; 11:e0152583. [PMID: 27028538 PMCID: PMC4814109 DOI: 10.1371/journal.pone.0152583] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/16/2016] [Indexed: 12/28/2022] Open
Abstract
Eukaryotic cells rapidly repair wounds on their plasma membrane. Resealing is Ca2+-dependent, and involves exocytosis of lysosomes followed by massive endocytosis. Extracellular activity of the lysosomal enzyme acid sphingomyelinase was previously shown to promote endocytosis and wound removal. However, whether lysosomal proteases released during cell injury participate in resealing is unknown. Here we show that lysosomal proteases regulate plasma membrane repair. Extracellular proteolysis is detected shortly after cell wounding, and inhibition of this process blocks repair. Conversely, surface protein degradation facilitates plasma membrane resealing. The abundant lysosomal cysteine proteases cathepsin B and L, known to proteolytically remodel the extracellular matrix, are rapidly released upon cell injury and are required for efficient plasma membrane repair. In contrast, inhibition of aspartyl proteases or RNAi-mediated silencing of the lysosomal aspartyl protease cathepsin D enhances resealing, an effect associated with the accumulation of active acid sphingomyelinase on the cell surface. Thus, secreted lysosomal cysteine proteases may promote repair by facilitating membrane access of lysosomal acid sphingomyelinase, which promotes wound removal and is subsequently downregulated extracellularly by a process involving cathepsin D.
Collapse
Affiliation(s)
- Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Christina Tam
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Norma W. Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
- * E-mail:
| |
Collapse
|
26
|
Liu Y, Zhu Z, Zhang M, Zheng H. Multifunctional roles of leader protein of foot-and-mouth disease viruses in suppressing host antiviral responses. Vet Res 2015; 46:127. [PMID: 26511922 PMCID: PMC4625562 DOI: 10.1186/s13567-015-0273-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/07/2015] [Indexed: 12/17/2022] Open
Abstract
Foot-and-mouth disease virus (FMDV) leader protein (Lpro) is a papain-like proteinase, which plays an important role in FMDV pathogenesis. Lpro exists as two forms, Lab and Lb, due to translation being initiated from two different start codons separated by 84 nucleotides. Lpro self-cleaves from the nascent viral polyprotein precursor as the first mature viral protein. In addition to its role as a viral proteinase, Lpro also has the ability to antagonize host antiviral effects. To promote FMDV replication, Lpro can suppress host antiviral responses by three different mechanisms: (1) cleavage of eukaryotic translation initiation factor 4 γ (eIF4G) to shut off host protein synthesis; (2) inhibition of host innate immune responses through restriction of interferon-α/β production; and (3) Lpro can also act as a deubiquitinase and catalyze deubiquitination of innate immune signaling molecules. In the light of recent functional and biochemical findings regarding Lpro, this review introduces the basic properties of Lpro and the mechanisms by which it antagonizes host antiviral responses.
Collapse
Affiliation(s)
- Yingqi Liu
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China. .,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Zixiang Zhu
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China.
| | - Miaotao Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China.
| |
Collapse
|
27
|
Birse K, Arnold KB, Novak RM, McCorrister S, Shaw S, Westmacott GR, Ball TB, Lauffenburger DA, Burgener A. Molecular Signatures of Immune Activation and Epithelial Barrier Remodeling Are Enhanced during the Luteal Phase of the Menstrual Cycle: Implications for HIV Susceptibility. J Virol 2015; 89:8793-805. [PMID: 26085144 PMCID: PMC4524071 DOI: 10.1128/jvi.00756-15] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/03/2015] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED The variable infectivity and transmissibility of HIV/SHIV has been recently associated with the menstrual cycle, with particular susceptibility observed during the luteal phase in nonhuman primate models and ex vivo human explant cultures, but the mechanism is poorly understood. Here, we performed an unbiased, mass spectrometry-based proteomic analysis to better understand the mucosal immunological processes underpinning this observed susceptibility to HIV infection. Cervicovaginal lavage samples (n = 19) were collected, characterized as follicular or luteal phase using days since last menstrual period, and analyzed by tandem mass spectrometry. Biological insights from these data were gained using a spectrum of computational methods, including hierarchical clustering, pathway analysis, gene set enrichment analysis, and partial least-squares discriminant analysis with LASSO feature selection. Of the 384 proteins identified, 43 were differentially abundant between phases (P < 0.05, ≥2-fold change). Cell-cell adhesion proteins and antiproteases were reduced, and leukocyte recruitment (interleukin-8 pathway, P = 1.41E-5) and extravasation proteins (P = 5.62E-4) were elevated during the luteal phase. LASSO/PLSDA identified a minimal profile of 18 proteins that best distinguished the luteal phase. This profile included cytoskeletal elements and proteases known to be involved in cellular movement. Gene set enrichment analysis associated CD4(+) T cell and neutrophil gene set signatures with the luteal phase (P < 0.05). Taken together, our findings indicate a strong association between proteins involved in tissue remodeling and leukocyte infiltration with the luteal phase, which may represent potential hormone-associated mechanisms of increased susceptibility to HIV. IMPORTANCE Recent studies have discovered an enhanced susceptibility to HIV infection during the progesterone-dominant luteal phase of the menstrual cycle. However, the mechanism responsible for this enhanced susceptibility has not yet been determined. Understanding the source of this vulnerability will be important for designing efficacious HIV prevention technologies for women. Furthermore, these findings may also be extrapolated to better understand the impact of exogenous hormone application, such as the use of hormonal contraceptives, on HIV acquisition risk. Hormonal contraceptives are the most widely used contraceptive method in sub-Saharan Africa, the most HIV-burdened area of the world. For this reason, research conducted to better understand how hormones impact host immunity and susceptibility factors important for HIV infection is a global health priority.
Collapse
Affiliation(s)
- Kenzie Birse
- National Lab for HIV Immunology, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kelly B Arnold
- Department of Biological Engineering and Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard M Novak
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Stuart McCorrister
- Mass Spectrometry Core Facility, National Microbiology Laboratory, Winnipeg, Manitoba, Canada
| | - Souradet Shaw
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Garrett R Westmacott
- Mass Spectrometry Core Facility, National Microbiology Laboratory, Winnipeg, Manitoba, Canada
| | - Terry B Ball
- National Lab for HIV Immunology, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada Department of Medical Microbiology, University of Nairobi, Department of Medical Microbiology, Nairobi, Kenya
| | - Douglas A Lauffenburger
- Department of Biological Engineering and Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Adam Burgener
- National Lab for HIV Immunology, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
Ferlizza E, Campos A, Neagu A, Cuoghi A, Bellei E, Monari E, Dondi F, Almeida A, Isani G. The effect of chronic kidney disease on the urine proteome in the domestic cat (Felis catus). Vet J 2015; 204:73-81. [DOI: 10.1016/j.tvjl.2015.01.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/20/2015] [Accepted: 01/24/2015] [Indexed: 01/29/2023]
|
29
|
Hoffman DR, Kroll LM, Basehoar A, Reece B, Cunningham CT, Koenig DW. Immediate and extended effects of abrasion on stratum corneum natural moisturizing factor. Skin Res Technol 2015; 21:366-72. [DOI: 10.1111/srt.12201] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2014] [Indexed: 12/31/2022]
Affiliation(s)
| | | | | | - B. Reece
- Reliance Clinical Testing Services, Inc.; Irving TX USA
| | | | | |
Collapse
|
30
|
Sage J, De Quéral D, Leblanc-Noblesse E, Kurfurst R, Schnebert S, Perrier E, Nizard C, Lalmanach G, Lecaille F. Differential expression of cathepsins K, S and V between young and aged Caucasian women skin epidermis. Matrix Biol 2014; 33:41-6. [PMID: 23871919 DOI: 10.1016/j.matbio.2013.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 10/26/2022]
Abstract
Cutaneous aging translates drastic structural and functional alterations in the extracellular matrix (ECM). Multiple mechanisms are involved, including changes in protease levels. We investigated the age-related protein expression and activity of cysteine cathepsins and the expression of two endogenous protein inhibitors in young and aged Caucasian women skin epidermis. Immunofluorescence studies indicate that the expression of cathepsins K, S and V, as well as cystatins A and M/E within keratinocytes is reduced in photoprotected skin of aged women. Furthermore, the overall endopeptidase activity of cysteine cathepsins in epidermis lysates decreased with age. Albeit dermal elastic fiber and laminin expression is reduced in aged skin, staining of nidogen-1, a key protein in BM assembly that is sensitive to proteolysis by cysteine, metallo- and serine proteases, has a similar pattern in both young and aged skin. Since cathepsins contribute to the hydrolysis and turnover of ECM/basement membrane components, the abnormal protein degradation and deposition during aging process may be related in part to a decline of lysosomal/endosomal cathepsin K, S and V activity.
Collapse
Affiliation(s)
- Juliette Sage
- INSERM, UMR 1100, Pathologies Respiratoires: protéolyse et aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France; LVMH-Recherche, BP58, F-45800 Saint Jean de Braye, France
| | | | | | - Robin Kurfurst
- LVMH-Recherche, BP58, F-45800 Saint Jean de Braye, France
| | | | - Eric Perrier
- LVMH-Recherche, BP58, F-45800 Saint Jean de Braye, France
| | - Carine Nizard
- LVMH-Recherche, BP58, F-45800 Saint Jean de Braye, France
| | - Gilles Lalmanach
- INSERM, UMR 1100, Pathologies Respiratoires: protéolyse et aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France
| | - Fabien Lecaille
- INSERM, UMR 1100, Pathologies Respiratoires: protéolyse et aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, F-37032 Tours cedex, France.
| |
Collapse
|
31
|
Hoffman DR, Kroll LM, Basehoar A, Reece B, Cunningham CT, Koenig DW. Immediate and extended effects of sodium lauryl sulphate exposure on stratum corneum natural moisturizing factor. Int J Cosmet Sci 2013; 36:93-101. [DOI: 10.1111/ics.12101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 10/15/2013] [Indexed: 12/19/2022]
Affiliation(s)
- D. R. Hoffman
- Kimberly-Clark Corporation; 2100 Winchester Road Neenah WI 54956 U.S.A
| | - L. M. Kroll
- Kimberly-Clark Corporation; 2100 Winchester Road Neenah WI 54956 U.S.A
| | - A. Basehoar
- Kimberly-Clark Corporation; 2100 Winchester Road Neenah WI 54956 U.S.A
| | - B. Reece
- Reliance Clinical Testing Services, Inc.; 3207 Esters Road Irving TX 75062-2879 U.S.A
| | - C. T. Cunningham
- Kimberly-Clark Corporation; 2100 Winchester Road Neenah WI 54956 U.S.A
| | - D. W. Koenig
- Kimberly-Clark Corporation; 2100 Winchester Road Neenah WI 54956 U.S.A
| |
Collapse
|
32
|
Current insights into protease dynamics in human epithelial disease and barrier function. Cell Tissue Res 2013; 351:213-5. [PMID: 23324990 DOI: 10.1007/s00441-013-1559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|