Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(12): e113956, 2014.
Article in English | MEDLINE | ID: mdl-25464502

ABSTRACT

Chronic and non-healing skin wounds represent a significant clinical, economic and social problem worldwide. Currently, there are few effective treatments. Lack of well-defined animal models to investigate wound healing mechanisms and furthermore to identify new and more effective therapeutic agents still remains a major challenge. Pig skin wound healing is close to humans. However, standardized pig wound healing models with demonstrated validity for testing new wound healing candidates are unavailable. Here we report a systematic evaluation and establishment of both acute and diabetic wound healing models in pigs, including wound-creating pattern for drug treatment versus control, measurements of diabetic parameters and the time for detecting delayed wound healing. We find that treatment and control wounds should be on the opposite and corresponding sides of a pig. We demonstrate a strong correlation between duration of diabetic conditions and the length of delay in wound closure. Using these new models, we narrow down the minimum therapeutic entity of secreted Hsp90α to a 27-amino acid peptide, called fragment-8 (F-8). In addition, results of histochemistry and immunohistochemistry analyses reveal more organized epidermis and dermis in Hsp90α-healed wounds than the control. Finally, Hsp90α uses a similar signaling mechanism to promote migration of isolated pig and human keratinocytes and dermal fibroblasts. This is the first report that shows standardized pig models for acute and diabetic wound healing studies and proves its usefulness with both an approved drug and a new therapeutic agent.


Subject(s)
Diabetes Complications , HSP90 Heat-Shock Proteins/therapeutic use , Models, Animal , Swine/physiology , Wound Healing/drug effects , Animals , Becaplermin , Cell Movement/drug effects , Diabetes Mellitus, Experimental/pathology , HSP90 Heat-Shock Proteins/pharmacology , HSP90 Heat-Shock Proteins/physiology , Proto-Oncogene Proteins c-sis/therapeutic use , Skin/injuries , Skin/metabolism , Skin/pathology , Time Factors , Wounds and Injuries/drug therapy
2.
Mol Cell Biol ; 33(24): 4947-59, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24126057

ABSTRACT

Normal cells secrete heat shock protein 90 alpha (Hsp90α) in response to tissue injury. Tumor cells have managed to constitutively secrete Hsp90α during invasion and metastasis. The sole function of extracellular Hsp90α (eHsp90α) is to promote cell motility, a critical event for both wound healing and tumor progression. The mechanism of promotility action by eHsp90α, however, has remained elusive. A key issue is whether eHsp90α still acts as a chaperone outside the cells or is a new and bona fide signaling molecule. Here, we have provided evidence that eHsp90α utilizes a unique transmembrane signaling mechanism to promote cell motility and wound healing. First, subdomain II in the extracellular part of low-density lipoprotein receptor-related protein 1 (LRP-1) receives the eHsp90α signal. Then, the NPVY but not the NPTY motif in the cytoplasmic tail of LRP-1 connects eHsp90α signaling to serine 473 but not threonine 308 phosphorylation in Akt kinases. Individual knockdown of Akt1, Akt2, or Akt3 revealed the importance of Akt1 and Akt2 in eHsp90α-induced cell motility. Akt gene rescue experiments suggest that Akt1 and Akt2 work in concert, rather than independently, to mediate eHsp90α promotility signaling. Finally, Akt1 and Akt2 knockout mice showed impaired wound healing that cannot be corrected by topical application with the eHsp90α protein.


Subject(s)
Cell Movement , HSP90 Heat-Shock Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Re-Epithelialization , Amino Acid Motifs , Animals , Cells, Cultured , Fibroblasts/physiology , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Mice, Knockout , Protein Structure, Tertiary , Signal Transduction , Skin/cytology
3.
Int Rev Cell Mol Biol ; 303: 203-35, 2013.
Article in English | MEDLINE | ID: mdl-23445811

ABSTRACT

Despite extensive investigative studies and clinical trials over the past two decades, we still do not understand why cancer cells are more sensitive to the cellular toxicity of Hsp90 inhibitors than normal cells. We still do not understand why only some cancer cells are sensitive to the Hsp90 inhibitors. Based on studies of the past few years, we argue that the selected sensitivity of cancer cells to Hsp90 inhibitors, such as 17-N-allylamino-17-demethoxygeldanamycin, is due to inhibition of the extracellular Hsp90 (eHsp90) rather than intracellular Hsp90 by these inhibitors. Because not all tumor cells utilize eHsp90 for motility, invasion and metastasis, only the group of "eHsp90-dependent" cancer cells is sensitive to Hsp90 inhibitors. If these notions prove to be true, pharmaceutical agents that selectively target eHsp90 should be more effective on tumor cells and less toxic on normal cells than current inhibitors that nondiscriminatively target both extracellular and intracellular Hsp90.


Subject(s)
Clinical Trials as Topic , Extracellular Space/metabolism , HSP90 Heat-Shock Proteins/metabolism , Blood Circulation , Cell Movement , Humans , Intracellular Space/metabolism
4.
Biochim Biophys Acta ; 1823(3): 730-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21982864

ABSTRACT

Extracellular Hsp90 proteins, including "membrane-bound", "released" and "secreted", were first reported more than two decades ago. Only studies of the past 7years have begun to reveal a picture for when, how and why Hsp90 gets exported by both normal and tumor cells. Normal cells secrete Hsp90 in response to tissue injury. Tumor cells have managed to constitutively secrete Hsp90 for tissue invasion. In either case, sufficient supply of the extracellular Hsp90 can be guaranteed by its unusually abundant storage inside the cells. A well-characterized function of secreted Hsp90α is to promote cell motility, a crucial event for both wound healing and cancer. The reported targets for extracellular Hsp90α include MMP2, LRP-1, tyrosine kinase receptors and possibly more. The pro-motility activity of secreted Hsp90α resides within a fragment, called 'F-5', at the boundary between linker region and middle domain. Inhibition of its secretion, neutralization of its extracellular action or interruption of its signaling through LRP-1 block wound healing and tumor invasion in vitro and in vivo. In normal tissue, topical application of F-5 promotes acute and diabetic wound healing far more effectively than US FDA-approved conventional growth factor therapy in mice. In cancer, drugs that selectively target the F-5 region of secreted Hsp90 by cancer cells may be more effective and less toxic than those that target the ATPase of the intracellular Hsp90. This article is part of a Special Issue entitled: Heat Shock Protein 90 (HSP90).


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Neoplasms/physiopathology , Wound Healing/physiology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Signal Transduction
5.
Mol Biol Cell ; 23(4): 602-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190738

ABSTRACT

Deregulated accumulation of hypoxia-inducible factor-1α (HIF-1α) is a hallmark of many solid tumors. Directly targeting HIF-1α for therapeutics is challenging. Our finding that HIF-1α regulates secretion of heat shock protein-90α (Hsp90α) for cell migration raises the exciting possibility that targeting the secreted Hsp90α from HIF-1α-positive tumors has a better clinical outlook. Using the HIF-1α-positive and metastatic breast cancer cells MDA-MB-231, we show that down-regulation of the deregulated HIF-1α blocks Hsp90α secretion and invasion of the cells. Reintroducing an active, but not an inactive, HIF-1α into endogenous HIF-1α-depleted cells rescues both Hsp90α secretion and invasion. Inhibition of Hsp90α secretion, neutralization of secreted Hsp90α action, or removal of the cell surface LRP-1 receptor for secreted Hsp90α reduces the tumor cell invasion in vitro and lung colonization and tumor formation in nude mice. Furthermore, we localized the tumor-promoting effect to a 115-amino acid region in secreted Hsp90α called F-5. Supplementation with F-5 is sufficient to bypass the blockade of HIF-1α depletion and resumes invasion by the tumor cells under serum-free conditions. Because normal cells do not secrete Hsp90α in the absence of stress, drugs that target F-5 should be more effective and less toxic in treatment of HIF-1α-positive tumors in humans.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/pathology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cell Line, Tumor , Cell Movement , Humans , Mice , Neoplasm Invasiveness , Neoplasms/metabolism , Peptides/pharmacology , RNA, Small Interfering/genetics
6.
J Clin Invest ; 121(11): 4348-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22019588

ABSTRACT

Wounds that fail to heal in a timely manner, for example, diabetic foot ulcers, pose a health, economic, and social problem worldwide. For decades, conventional wisdom has pointed to growth factors as the main driving force of wound healing; thus, growth factors have become the center of therapeutic developments. To date, becaplermin (recombinant human PDGF-BB) is the only US FDA-approved growth factor therapy, and it shows modest efficacy, is costly, and has the potential to cause cancer in patients. Other molecules that drive wound healing have therefore been sought. In this context, it has been noticed that wounds do not heal without the participation of secreted Hsp90α. Here, we report that a 115-aa fragment of secreted Hsp90α (F-5) acts as an unconventional wound healing agent in mice. Topical application of F-5 peptide promoted acute and diabetic wound closure in mice far more effectively than did PDGF-BB. The stronger effect of F-5 was due to 3 properties not held by conventional growth factors: its ability to recruit both epidermal and dermal cells; the fact that its ability to promote dermal cell migration was not inhibited by TGF-ß; and its ability to override the inhibitory effects of hyperglycemia on cell migration in diabetes. The discovery of F-5 challenges the long-standing paradigm of wound healing factors and reveals a potentially more effective and safer agent for healing acute and diabetic wounds.


Subject(s)
HSP90 Heat-Shock Proteins/pharmacology , Wound Healing/drug effects , Administration, Topical , Animals , Becaplermin , Cells, Cultured , Diabetes Complications/drug therapy , Diabetes Complications/physiopathology , HSP90 Heat-Shock Proteins/administration & dosage , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/physiology , Humans , Mice , Mice, Hairless , Mice, Nude , Models, Biological , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Wound Healing/physiology
7.
Cell Microbiol ; 5(7): 445-53, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12814435

ABSTRACT

We have assessed the kinetics of host gene expression in granulomas of mice infected with virulent Mycobacterium tuberculosis, using an approach that incorporates the laser capture microdissection (LCM) and real-time PCR technology in conjunction with a newly derived mathematical equation. The results have provided evidence indicating that conventional use of whole infected lungs to study granuloma-specific gene expression can yield data that may not genuinely reflect intralesional events. Significantly, the expression of nine host genes known to regulate the inflammatory response to M. tuberculosis, as determined by real-time PCR analysis of microdissected granuloma-derived cDNAs, was downregulated (up to 27-fold) at around the time when the rapid growth phase of the bacilli in the lungs of infected mice ends. This downregulation was masked when whole infected lungs were used for the studies. The data suggest that the host immune system can adjust and respond to, or can be modulated by specific physiological states of the tubercle bacillus in vivo. The LCM/real-time PCR-based system described in this study can be applied to safely and accurately evaluate gene expression in any lesions that can be microscopically visualized, including those contained in biohazardous tissues.


Subject(s)
Gene Expression , Tuberculoma/genetics , Tuberculoma/immunology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Animals , Down-Regulation , Female , Gene Expression Profiling , Lasers , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Micromanipulation , Mycobacterium tuberculosis/pathogenicity , Polymerase Chain Reaction , Proteins/genetics , Proteins/metabolism , Tuberculoma/microbiology , Tuberculosis, Pulmonary/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...