Network


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

Hotspot


Dive into the research topics where Kathleen C. Flanders is active.

Publication


Featured researches published by Kathleen C. Flanders.


Cell | 1987

Evidence that transforming growth factor-β is a hormonally regulated negative growth factor in human breast cancer cells

Cornelius Knabbe; Marc E. Lippman; Lalage M. Wakefield; Kathleen C. Flanders; Attan Kasid; Rik Derynck; Robert B. Dickson

The hormone-dependent human breast cancer cell line MCF-7 secretes transforming growth factor-beta (TGF-beta), which can be detected in the culture medium in a biologically active form. These polypeptides compete with human platelet-derived TGF-beta for binding to its receptor, are biologically active in TGF-beta-specific growth assays, and are recognized and inactivated by TGF-beta-specific antibodies. Secretion of active TGF-beta is induced 8 to 27-fold under treatment of MCF-7 cells with growth inhibitory concentrations of antiestrogens. Antiestrogen-induced TGF-beta from MCF-7 cells inhibits the growth of an estrogen receptor-negative human breast cancer cell line in coculture experiments; growth inhibition is reversed with anti-TGF-beta antibodies. We conclude that in MCF-7 cells, TGF-beta is a hormonally regulated growth inhibitor with possible autocrine and paracrine functions in breast cancer cells.


Annals of the New York Academy of Sciences | 1990

Transforming Growth Factor-β

Anita B. Roberts; Ursula I. Heine; Kathleen C. Flanders; Michael B. Sporn

Transforming growth factor-β (TGF-β) is generally acknowledged to be the cytokine with the broadest range of activities in repair of injured tissue, based both on the variety of cell types that produce and/or respond to it and on the spectrum of its cellular responses (Roberts and Sporn, 1990). TGF-β is released from degranulating platelets and secreted by all of the major cell types participating in the repair process, including lymphocytes, macrophages, endothelial cells, smooth muscle cells, epithelial cells, and fibroblasts (see Fig. 1). A unique feature of this molecule is that its autoinduction results in sustained expression at the site of a wound and extends the effectiveness of both the initial burst of endogenous TGF-β released upon injury and exogenous TGF-β that might be applied to a wound. The ability of TGF-β to improve and/or accelerate tissue repair has been studied extensively in a variety of animal models of both normal and impaired healing. A limited number of clinical trials are in progress, but it is anticipated that many new applications for TGF-β will ultimately be found, once problems with appropriate timing and formulation can be solved.


American Journal of Pathology | 2002

Mice Lacking Smad3 Are Protected Against Cutaneous Injury Induced by Ionizing Radiation

Kathleen C. Flanders; Catherine D. Sullivan; Makiko Fujii; Anastasia L. Sowers; Mario A. Anzano; Alidad Arabshahi; Christopher Major; Chu-Xia Deng; Angelo Russo; James B. Mitchell; Anita B. Roberts

Transforming growth factor-beta (TGF-beta) plays a central role in the pathogenesis of inflammatory and fibrotic diseases, including radiation-induced fibrosis. We previously reported that mice null for Smad3, a key downstream mediator of TGF-beta, show accelerated healing of cutaneous incisional wounds with reduced inflammation and accumulation of matrix. To determine if loss of Smad3 decreases radiation-induced injury, skin of Smad3+/+ [wild-type (WT)] and -/- [knockout (KO)] mice was exposed to a single dose of 30 to 50 Gy of gamma-irradiation. Six weeks later, skin from KO mice showed significantly less epidermal acanthosis and dermal influx of mast cells, macrophages, and neutrophils than skin from WT littermates. Skin from irradiated KO mice exhibited less immunoreactive TGF-beta and fewer myofibroblasts, suggesting that these mice will have a significantly reduced fibrotic response. Although irradiation induced no change in the immunohistochemical expression of the TGF-beta type I receptor, the epidermal expression of the type II receptor was lost after irradiation whereas its dermal expression remained high. Primary keratinocytes and dermal fibroblasts prepared from WT and KO mice showed similar survival when irradiated, as did mice exposed to whole-body irradiation. These results suggest that inhibition of Smad3 might decrease tissue damage and reduce fibrosis after exposure to ionizing irradiation.


Molecular Cancer Therapeutics | 2007

Pharmacodynamic characterization of chemopreventive triterpenoids as exceptionally potent inducers of Nrf2-regulated genes

Melinda S. Yates; Masafumi Tauchi; Fumiki Katsuoka; Kathleen C. Flanders; Karen T. Liby; Tadashi Honda; Gordon W. Gribble; Delinda A. Johnson; Jeffrey A. Johnson; Neal C. Burton; Tomás R. Guilarte; Masayuki Yamamoto; Michael B. Sporn; Thomas W. Kensler

Synthetic triterpenoids have been developed, which are potent inducers of cytoprotective enzymes and inhibitors of inflammation, greatly improving on the weak activity of naturally occurring triterpenoids. An imidazolide triterpenoid derivative, 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im or TP235), has been previously shown to potently protect against hepatic tumorigenesis, acting in part by inducing cytoprotective genes through Keap1-Nrf2-antioxidant response element (ARE) signaling. In these studies, the pharmacodynamic activity of CDDO-Im is characterized in two distinct lines of ARE reporter mice and by measuring increases in Nqo1 transcript levels as a marker of cytoprotective gene induction. Oral administration of CDDO-Im induces ARE-regulated cytoprotective genes in many tissues in the mouse, including liver, lung, kidney, intestines, brain, heart, thymus, and salivary gland. CDDO-Im induces Nqo1 RNA transcripts in some organs at doses as low as 0.3 μmol/kg body weight (orally). A structure activity evaluation of 15 additional triterpenoids (a) confirmed the importance of Michael acceptor groups on both the A and C rings, (b) showed the requirement for a nitrile group at C-2 of the A ring, and (c) indicated that substituents at C-17 dramatically affected pharmacodynamic action in vivo. In addition to CDDO-Im, other triterpenoids, particularly the methyl ester CDDO-Me (TP155) and the dinitrile TP225, are extremely potent inducers of cytoprotective genes in mouse liver, lung, small intestine mucosa, and cerebral cortex. This pharmacodynamic characterization highlights the chemopreventive promise of several synthetic triterpenoids in multiple target organs. [Mol Cancer Ther 2007;6(1):154–62]


American Journal of Pathology | 2004

Smad3 Signaling Is Required for Epithelial-Mesenchymal Transition of Lens Epithelium after Injury

Shizuya Saika; Satoko Kono-Saika; Yoshitaka Ohnishi; Misako Sato; Yasuteru Muragaki; Akira Ooshima; Kathleen C. Flanders; Jiyun Yoo; Mario A. Anzano; Chia-Yang Liu; Winston W.-Y. Kao; Anita B. Roberts

Lens epithelial cells undergo epithelial-mesenchymal transition (EMT) after injury as in cataract extraction, leading to fibrosis of the lens capsule. Fibrosis of the anterior capsule can be modeled in the mouse by capsular injury in the lens, which results in EMT of the lens epithelium and subsequent deposition of extracellular matrix without contamination of other cell types from outside the lens. We have previously shown that signaling via Smad3, a key signal-transducing element downstream of transforming growth factor (TGF)-beta and activin receptors, is activated in lens epithelial cells by 12 hours after injury and that this Smad3 activation is blocked by administration of a TGF-beta 2-neutralizing antibody in mice. We now show that EMT of primary lens epithelial cells in vitro depends on TGF-beta expression and that injury-induced EMT in vivo depends, more specifically, on signaling via Smad3. Loss of Smad3 in mice blocks both morphological changes of lens epithelium to a mesenchymal phenotype and expression of the EMT markers snail, alpha-smooth muscle actin, lumican, and type I collagen in response to injury in vivo or to exposure to exogenous TGF-beta in organ culture. The results suggest that blocking the Smad3 pathway might be beneficial in inhibiting capsular fibrosis after injury and/or surgery.


Recent Progress in Hormone Research | 1988

Transforming growth factor beta: biochemistry and roles in embryogenesis, tissue repair and remodeling, and carcinogenesis.

Anita B. Roberts; Kathleen C. Flanders; Paturu Kondaiah; Nancy L. Thompson; Ellen Van Obberghen-Schilling; Lalage M. Wakefield; Pellegrino Rossi; Benoit de Crombrugghe; Ursula I. Heine; Michael B. Sporn

Transforming growth factor (TGF)- β plays essential roles in embryogenesis, particularly during periods of morphogenesis. Some of the same embryological mechanisms are reiterated in the adult during the normal processes of tissue remodeling and repair and aberrantly in various pathological processes, including carcinogenesis. This chapter highlights the new advances in the understanding of the complex biology of TGF- β and discusses the chemistry of TGF- β . The broad range of biological activities of TGF- β makes it highly likely that other peptide activities—purified by presumably novel and specific assays—will result from TGF- β once their amino acid sequence is determined. TGF- β 1 and 2 are two homologous forms of a homodimeric peptide with molecular weight of 25,000. Every chain of the peptide contains 112 amino acids of which nine are cysteine residues. The chapter reviews the structure of TGF- β 1 and 2 and TGF- β gene family. The biological activities of the members of the TGF- β family are described in the chapter. The chapter further reviews the regulation of gene activity by TGF- β, antibodies to TGF- β , and role of TGF- β in embryogenesis, tissue repair and remodeling, and carcinogenesis and other proliferative diseases.


Growth Factors Journal | 1988

Transforming Growth Factor Beta-1 in Acute Myocardial Infarction in Rats

Nancy L. Thompson; Fernando Bazoberry; Edith Speir; Ward Casscells; Victor J. Ferrans; Kathleen C. Flanders; Patljru Kondaiah; Andrew G. Geiser; Michael B. Sporn

TGF-beta 1 has been examined in the heart during myocardial infarction caused by ligation of the left coronary artery. Infarcted and uninfarcted myocardium have been compared by immunohistochemical staining of TGF-beta 1 and by Northern blot analysis of mRNA. Normal ventricular myocytes are strongly stained by an antibody to TGF-beta 1. Progressive loss of staining of these myocytes begins within 1 hr after coronary ligation. However, by 24-48 hr after ligation, intense staining of myocytes at the margin of infarcted areas is seen. Northern blots of infarcted myocardium 48 hr after ligation show a 3- to 4-fold increase in the principal 2.4 kb TGF-beta 1 mRNA; there is also a marked increase in a minor 1.9 kb transcript. In the same tissue samples, there is a 2-fold decrease in the mRNA for the glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase. The results indicate a significant role for TGF-beta in the response of the heart to injury.


Annals of the New York Academy of Sciences | 2003

Smad3: A Key Player in Pathogenetic Mechanisms Dependent on TGF‐β

Anita B. Roberts; Angelo Russo; Angelina Felici; Kathleen C. Flanders

Abstract: Transforming growth factor‐β (TGF‐β), a key player in a large variety of physiological and disease processes, signals through transmembrane receptor serine/threonine kinases to activate novel signaling intermediates called Smad proteins, which then modulate transcription of target genes. We have utilized mice with a targeted deletion of Smad3, one of two homologous proteins involved in signaling from TGF‐β/activin, to investigate the function of this particular pathway in transducing such effects of TGF‐β. The dramatic results of the absence of Smad3 on parameters of healing of cutaneous wounds, such as reepithelialization and influx of inflammatory cells, as well as on fibrosis as modeled by radiation fibrosis of skin in mice, suggest that signaling flux through Smad3 is critical for chemotactic activity of TGF‐β, inhibitory effects of TGF‐β on keratinocyte proliferation and migration, and chemoattraction and elaboration of extracellular matrix by fibroblasts in fibrotic diseases. We recently identified a novel molecule, TLP for TRAP‐1‐like protein, which selectively interferes with Smad3 signaling, and are currently investigating whether levels of this protein might be altered in disease to change the relative flow of information from Smad2 and Smad3.


Human Pathology | 1993

Association of transforming growth factor-β1 with prostate cancer: An immunohistochemical study

Luan D. Truong; Dov Kadmon; Bryan K. McCune; Kathleen C. Flanders; Peter T. Scardino; Timothy C. Thompson

Prostate tissue samples from patients with prostatic carcinoma (PC) and/or benign prostatic hyperplasia (BPH) were examined for expression of transforming growth factor-beta 1 (TGF-beta 1) using an immunohistochemical technique. Tissues were stained with CC and LC antisera, which react with extracellular and intracellular TGF-beta 1, respectively. All PC and BPH tissues showed positive extracellular staining; however, CC-immunoreactive material was significantly more extensive in PC compared with BPH, the average positively staining areas being 59% and 26%, respectively. This differential staining pattern was evident in cases in which areas of PC were located adjacent to areas of BPH. LC staining was identified exclusively intracellularly involving both stromal and epithelial cells in cases of PC as well as BPH. However, while stromal cell staining was more pronounced in BPH, epithelial cell staining tended to be more extensive and more intense in PC. The findings suggest that TGF-beta 1 may be biologically important in the development of PC and BPH.


American Journal of Pathology | 2003

Interference with Transforming Growth Factor-β/ Smad3 Signaling Results in Accelerated Healing of Wounds in Previously Irradiated Skin

Kathleen C. Flanders; Christopher Major; Alidad Arabshahi; Ekinadese E. Aburime; Miya H. Okada; Makiko Fujii; Timothy D. Blalock; Gregory S. Schultz; Anastasia L. Sowers; Mario A. Anzano; James B. Mitchell; Angelo Russo; Anita B. Roberts

Transforming growth factor (TGF)-beta regulates many aspects of wound repair including inflammation, chemotaxis, and deposition of extracellular matrix. We previously showed that epithelialization of incisional wounds is accelerated in mice null for Smad3, a key cytoplasmic mediator of TGF-beta signaling. Here, we investigated the effects of loss of Smad3 on healing of wounds in skin previously exposed to ionizing radiation, in which scarring fibrosis complicates healing. Cutaneous wounds made in Smad3-null mice 6 weeks after irradiation showed decreased wound widths, enhanced epithelialization, and reduced numbers of neutrophils and myofibroblasts compared to wounds in irradiated wild-type littermates. Differences in breaking strength of wild-type and Smad3-null wounds were not significant. As shown previously for neutrophils, chemotaxis of primary dermal fibroblasts to TGF-beta required Smad3, but differentiation of fibroblasts to myofibroblasts by TGF-beta was independent of Smad3. Previous irradiation-enhanced induction of connective tissue growth factor mRNA in wild-type, but not Smad3-null fibroblasts, suggested that this may contribute to the heightened scarring in irradiated wild-type skin as demonstrated by Picrosirius red staining. Overall, the data suggest that attenuation of Smad3 signaling might improve the healing of wounds in previously irradiated skin commensurate with an inhibition of fibrosis.

Collaboration


Dive into the Kathleen C. Flanders's collaboration.

Top Co-Authors

Avatar

Anita B. Roberts

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lalage M. Wakefield

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shizuya Saika

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar

Osamu Yamanaka

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar

Akira Ooshima

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar

Yoshitaka Ohnishi

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuka Okada

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge