Network


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

Hotspot


Dive into the research topics where Thomas M. Walz is active.

Publication


Featured researches published by Thomas M. Walz.


International Archives of Allergy and Immunology | 1997

Localization of granule proteins in human eosinophil bone marrow progenitors.

Arne Egesten; Jero Calafat; Peter F. Weller; Edward F. Knol; Hans Janssen; Thomas M. Walz; Inge Olsson

Eosinophils have a characteristic content of cationic proteins, stored in core-containing specific granules and released at sites of inflammation; coreless granules (sometimes called primary) are present in eosinophil promyelocytes. In order to determine a possible relationship between the two granule subsets, immunoelectron-microscopic techniques were used to determine the presence and precise intragranular distribution of major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil peroxidase (EPO), and arylsulfatase B of eosinophil granules, as well as the Charcot-Leyden crystal (CLC) protein, in eosinophil progenitors of the bone marrow. MBP, ECP, EPO, and arylsulfatase B were observed in both coreless and core-containing (specific) granules. The difference in the distribution of MBP, having a uniform distribution in coreless granules and a crystalloid distribution in core-containing (specific) granules, could indicate a maturational process of a common organelle. CLC protein was distributed in the cytosol, in the euchromatin of the nuclei, but was also present in a rare granular compartment of both immature and mature eosinophils. The present findings suggest that coreless granules develop into core-containing specific granules.


Experimental Cell Research | 1992

α-Smooth muscle actin is expressed in a subpopulation of cultured and cloned fibroblasts and is modulated by γ-interferon

Alexis Desmoulière; Laura Rubbia-Brandt; Avni Abdiu; Thomas M. Walz; Alvaro Macieira-Coelho; Giulio Gabbiani

Clinical and experimental investigations have shown that, during wound healing and fibrocontractive diseases, fibroblasts acquire, more or less permanently according to the situation, morphological and biochemical features of smooth muscle (SM) cells including the expression of alpha-SM actin. Primary and passaged cultures of rat and human fibroblasts contain a subpopulation of cells expressing alpha-SM actin. These cells could derive from SM cells and/or pericytes present in the tissue from which cultures have been produced or represent bona fide fibroblasts. We have investigated the presence of alpha-SM actin in fibroblast cultures, clones, and subclones. In all cases the fibroblastic populations studied showed a proportion of alpha-SM actin expressing cells. Even after cloning, we never obtained populations negative for alpha-SM actin. We conclude that alpha-SM actin expression in fibroblastic cultures is not due to contaminant cells but is a feature of fibroblasts themselves. Our results support the view that fibroblastic cells are a heterogeneous population. It has been previously shown that gamma-interferon (gamma-IFN) decreases alpha-SM actin expression in SM cells. In rat and human fibroblasts, gamma-IFN decreases alpha-SM actin protein and mRNA expression as well as proliferation. The properties of this cytokine make it a good candidate for exerting an anti-fibrotic activity in vivo.


Scandinavian Journal of Rheumatology | 2005

TGF-alpha and ErbB2 production in synovial joint tissue: increased expression in arthritic joints.

Anna-Lotta Hallbeck; Thomas M. Walz; Kristina Briheim; Åke Wasteson

Objective: Cell types present in synovial joint tissues and during synovitis are known to produce epidermal growth factor receptor (EGFR)/ErbB‐1/HER‐1 and the potent EGFR‐ligand transforming growth factor‐alpha (TGF‐α) in vitro. Concomitant expression of TGF‐α, EGFR, and ErbB2 gives a strong proliferative drive in vitro and in vivo. However, the presence of TGF‐α and members of the EGFR/EGFR‐ligand family has not been thoroughly investigated in joint tissue in vivo. We aimed to determine whether TGF‐α, EGFR, and ErbB2 are present in human synovial joints, especially during rheumatoid arthritis (RA). Methods: TGF‐α protein was immunodetected in knee synovial fluid (SF) collected from 23 RA patients, eight patients with other arthritic conditions, two osteoarthritis (OA) patients, and six post‐traumatic patients (control). TGF‐α mRNA and TGF‐α, ErbB2, EGFR, and CD68 immunoreactivity were detected in knee synovial biopsies (6 RA/2 OA/6 control) using in situ hybridization and immunohistochemistry. TGF‐α mRNA was determined in SF cells by reverse transcription polymerase chain reaction (RT‐PCR) and/or the Northern blot technique. Results: TGF‐α protein was found in the synovial membrane (SM) and in the majority of SF samples. TGF‐α levels were significantly higher (p<0.001) in SF of RA patients than controls, TGF‐α protein and mRNA were increased and more widespread in SM of RA patients. In addition, white blood cells collected from RA SF expressed TGF‐α mRNA. Immunoreactivity for ErbB2 was found in SM and was more widespread in RA patients than in controls. Conclusion: The presence of TGF‐α in normal SF and SM may indicate a physiological maintenance function. The increased expression of TGF‐α and ErbB2 in RA SF and SM may give rise to an abnormal growth pattern, contributing to inflammatory synovial hyperplasia.


Melanoma Research | 2009

ErbB receptor tyrosine kinases contribute to proliferation of malignant melanoma cells : inhibition by gefitinib (ZD1839)

Emelie A. Djerf; Cecilia Trinks; Avni Abdiu; Lena K. Thunell; Anna-Lotta Hallbeck; Thomas M. Walz

Members of the epidermal growth factor (EGF) family of structurally related tyrosine kinase receptors, known as the ErbB receptors (EGFR/ErbB1/HER1, ErbB2/HER2/neu, ErbB3/HER3 and ErbB4/HER4) and their respective ligands, have been suggested to be involved in the development and progression of malignant melanoma. Here we investigate the effects of the ErbB1 tyrosine kinase inhibitor gefitinib (ZD1839, Iressa) on human malignant melanoma cells (RaH3 and RaH5) in vitro. ZD1839 inhibited proliferation of exponentially growing RaH3 and RaH5 cells in a dose-dependent manner with a half-maximally effective dose of 3.5 and 2.0u2009μmol/l, respectively. Cell growth was inhibited at 0.1u2009μmol/l ZD1839 in both cell lines. Maximal inhibition was accomplished at 10u2009μmol/l ZD1839; however, the effect was not complete as both cell lines showed a continuous slow growth during the treatment period. Flow cytometry analysis of cell-cycle distribution showed that ZD1839 treatment caused accumulation of RaH3 and RaH5 cells in the G1 phase. The growth arrest induced by ZD1839 coincided with upregulation of the cyclin-dependent kinase inhibitor p27KIP1. There was no increase in apoptosis as determined by analysis of plasma phosphatidyl serine redistribution. Western blot analysis revealed that ZD1839 substantially reduced tyrosine phosphorylation of ErbB1 as well as ErbB2 and ErbB3. This was accompanied by a concomitant decrease in Akt-phosphorylation, Erk1/2-phosphorylation, and Stat3-phosphorylation. Our results show that ZD1839 interferes with the growth of human malignant melanoma cells by cytostatic effects. These findings indicate the possible use of ErbB receptor kinase inhibitors as a novel treatment strategy in malignant melanoma.


Breast Cancer Research and Treatment | 2012

Individually tailored treatment with epirubicin and paclitaxel with or without capecitabine as first-line chemotherapy in metastatic breast cancer: a randomized multicenter trial

Thomas Hatschek; Lena Carlsson; Zakaria Einbeigi; Elisabet Lidbrink; Barbro Linderholm; Birgitta Lindh; Niklas Loman; Martin Malmberg; Samuel Rotstein; Martin Söderberg; Marie Sundquist; Thomas M. Walz; Mats Hellström; Helene Svensson; G. Astrom; Yvonne Brandberg; John Carstensen; Mårten Fernö; Jonas Bergh

Anthracyclines and taxanes are active cytotoxic drugs in the treatment of early metastatic breast cancer. It is yet unclear whether addition of capecitabine to the combination of these drugs improves the treatment outcome. Patients with advanced breast cancer were randomized to first-line chemotherapy with a combination of epirubicin (Farmorubicin®) and paclitaxel (Taxol®) alone (ET) or in combination with capecitabine (Xeloda®, TEX). Starting doses for ET were epirubicin 75xa0mg/m2 plus paclitaxel 175xa0mg/m2, and for TEX epirubicin 75xa0mg/m2, paclitaxel 155xa0mg/m2, and capecitabine 825xa0mg/m2 BID for 14xa0days. Subsequently, doses were tailored related to side effects. Primary endpoint was progression-free survival (PFS); secondary endpoints were overall survival (OS), time to treatment failure (TTF), objective response (OR), safety and quality of life (QoL). 287 patients were randomized, 143 to ET and 144 to TEX. Median PFS was 10.8xa0months for patients treated with ET, and 12.4xa0months for those treated with TEX (HR 0.84, 95% CI 0.65–1.07, Pxa0=xa00.16); median OS was 26.0xa0months for women in the ET versus 29.7xa0months in the TEX arm (HR 0.84, 95% CI 0.63–1.11, Pxa0=xa00.22). OR was achieved in 44.8% (ET) and 54.2% (TEX), respectively (χ2 3.66, Pxa0=xa00.16). TTF was significantly longer for patients treated with TEX, 6.0xa0months, versus 5.2xa0months following ET (HR 0.73, 95% CI 0.58–0.93, Pxa0=xa00.009). Severe hematological side effects related to epirubicin and paclitaxel were evenly distributed between the treatment arms, mucositis, diarrhea, and Hand-Foot syndrome were significantly more frequent in the TEX arm. Toxicity-adjusted treatment with ET and TEX showed similar efficacy in terms of PFS, OS, and OR. In this trial with limited power, the addition of capecitabine to epirubicin and paclitaxel as first-line treatment did not translate into clinically relevant improvement of the outcome.


Biochemical and Biophysical Research Communications | 2011

The pan-ErbB receptor tyrosine kinase inhibitor canertinib promotes apoptosis of malignant melanoma in vitro and displays anti-tumor activity in vivo

Emelie Severinsson; Cecilia Trinks; Henrik Gréen; Avni Abdiu; Anna-Lotta Hallbeck; Olle Stål; Thomas M. Walz

The ErbB receptor family has been suggested to constitute a therapeutic target for tumor-specific treatment of malignant melanoma. Here we investigate the effect of the pan-ErbB tyrosine kinase inhibitor canertinib on cell growth and survival in human melanoma cells in vitro and in vivo. Canertinib significantly inhibited growth of cultured melanoma cells, RaH3 and RaH5, in a dose-dependent manner as determined by cell counting. Half-maximum growth inhibitory dose (IC(50)) was approximately 0.8 μM and by 5 μM both cell lines were completely growth-arrested within 72 h of treatment. Incubation of exponentially growing RaH3 and RaH5 with 1 μM canertinib accumulated the cells in the G(1)-phase of the cell cycle within 24h of treatment without induction of apoptosis as determined by flow cytometry. Immunoblot analysis showed that 1 μM canertinib inhibited ErbB1-3 receptor phosphorylation with a concomitant decrease of Akt-, Erk1/2- and Stat3 activity in both cell lines. In contrast to the cytostatic effect observed at doses ≤ 5μM canertinib, higher concentrations induced apoptosis as demonstrated by the Annexin V method and Western blot analysis of PARP cleavage. Furthermore, canertinib significantly inhibited growth of RaH3 and RaH5 melanoma xenografts in nude mice. Pharmacological targeting of the ErbB receptors may prove successful in the treatment of patients with metastatic melanoma.


Biochemical and Biophysical Research Communications | 2010

The pan-ErbB receptor tyrosine kinase inhibitor canertinib induces ErbB-independent apoptosis in human leukemia (HL-60 and U-937) cells

Cecilia Trinks; Emelie A. Djerf; Anna-Lotta Hallbeck; Jan-Ingvar Jönsson; Thomas M. Walz

Epidermal growth factor (EGF) receptor tyrosine kinase inhibitors have recently been shown to display anti-neoplastic effects in human malignant myeloid cells. Our study was initiated in order to determine the effect of the pan-ErbB receptor tyrosine kinase inhibitor, canertinib (CI-1033), on growth and survival of human leukemia (HL-60 and U-937) cells. We show that treatment of HL-60 and U-937 cells with canertinib significantly inhibits growth of both cell lines in a dose-dependent manner; half maximal effective dose (IC(50)) in HL-60 and U-937 cells was approximately 2.5 microM and 1.0 microM, respectively. Treatment with 2 microM canertinib promoted a G(1) cell cycle arrest, whereas doses of 5 microM or more induced apoptosis as determined by the Annexin V method and cleavage of poly-(ADP-ribose) polymerase (PARP). HL-60 and U-937 cells lacked EGF-receptor transcript but expressed ErbB2-4 mRNA as determined by RT-PCR. However, none of the corresponding ErbB-receptor proteins could be detected by Western blot analysis. We conclude that canertinib induces apoptosis in HL-60 and U-937 cells devoid of functional ErbB1-4 receptors. Our results suggest that canertinib could be of potential clinical interest in the treatment of acute myeloid leukemia.


Bioscience Reports | 2001

Interleukin-6 Enhances Transforming Growth Factor-alpha mRNA Expression in Macrophage-Like Human Monocytoid (U-937-1) Cells

Anna-Lotta Hallbeck; Thomas M. Walz; Åke Wasteson

We have previously reported that the human monocytoid cell line U-937-1 constitutively expresses transforming growth factor-alpha (TGF-α) and that the steady-state levels of TGF-α mRNA as well as TGF-α protein release increase when U-937-1 cells are differentiated towards monocytes/macrophages. Interleukin-6 (IL-6), which has been shown to have growth-stimulatory effects on a number of cell types, has recently been shown to enhance TGF-α expression in keratinocytes. In the present study we investigated whether TGF-α expression in macrophage-like cells could be regulated by IL-6 using U-937-1 cells as a model system of monocyte/macrophage differentiation.U-937-1 cells were differentiated with retinoic acid (RA), vitamin D3 (Vit-D3) or phorbol-12-myristate-13-acetate (PMA) for 4 days and were then treated with human recombinant IL-6 (1000 IU/ml) for up to 24 hr. Northern blot analysis revealed that cells differentiated with PMA, inducing the phenotype of a secretory macrophage, markedly increased their TGF-α mRNA levels (2.7-fold) when treated with IL-6; the response was maximal at 6 hr and remained high at 12 hr. The expression of the TGF-α gene was accompanied by release of TGF-α protein into the cell culture medium, irrespective of differentiating agent, as demonstrated by enzyme-linked immunosorbent assay (ELISA), as well as by surface expression of pro-TGF-α as determined by indirect immunofluorescent cytometry. However, the superinduction of the TGF-α gene by IL-6 in cells differentiated with PMA was not accompanied by any increase in TGF-α protein release or pro-TGF-α surface expression.We conclude that since IL-6 causes increased steady-state levels of TGF-α mRNA in macrophage-like cells, it may prime these cells for production of this growth factor. Furthermore, we have shown that the IL-6 receptor complex is functional in U-937-1 cells induced to differentiate towards a secretory macrophage by treatment with PMA.


Cancer Letters | 1999

Suramin blocks growth-stimulatory effects of platelet-derived growth factor on malignant fibrous histiocytomas in vitro

Avni Abdiu; Sven-Erik Larsson; Åke Wasteson; Thomas M. Walz

The pattern of susceptibility of malignant cells to the cytostatic drug suramin is not fully clarified. Therefore, in the present paper we have assessed the effects of suramin on the growth of eight cell lines derived from human malignant fibrous histiocytomas, by measuring DNA synthesis. The effect of suramin (10-200 microg/ml) on cells either unstimulated, or stimulated with platelet-derived growth factor (PDGF)-AA (10 ng/ml), PDGF-BB (10 ng/ml) or 10% fetal calf serum was studied. Four out of five cell lines unable to thrive without external growth factors showed growth inhibition by suramin. The two cell lines able to grow under serum-free conditions were unaffected by high-dose suramin. The exposure to suramin, at 200 microg/ml, abolished the growth stimulation caused by PDGF-AA and -BB. In contrast, a low dose of suramin (50 microg/ml), with or without PDGF, caused growth-stimulating effects in some cell lines. Our results indicate that high doses of suramin inhibit growth of malignant fibrous histiocytomas in vitro and that suramin exerts its growth-inhibitory effects on cells dependent on external growth factors. Low-dose treatment with suramin, however, may instead promote growth in both serum-dependent and -independent tumor cell lines.


Cancer Letters | 1999

Effects of human platelet-derived growth factor-AB on sarcoma growth in vitro and in vivo

Avni Abdiu; Sten Wingren; Sven-Erik Larsson; Åke Wasteson; Thomas M. Walz

Platelet-derived growth factor (PDGF) has been proposed to play an important role in the growth of tumors. In order to study the effects of PDGF-AB on tumor growth in vivo, sarcoma-bearing mice were treated with PDGF-AB. The tumors, a malignant fibrous histiocytoma and an osteosarcoma, had functional PDGF receptors in vitro, as demonstrated by stimulation of PDGF-AB using a [3H]thymidine incorporation assay. Immunohistochemistry also revealed that both sarcoma xenografts expressed PDGF receptors. The tumor-bearing mice were given human PDGF-AB for 14 days, either continuously by an intraperitoneally placed mini-osmotic pump, or by daily injections. No effects on tumor growth in vivo were observed, as measured by tumor volume, autoradiography or cell cycle distribution. The histological appearance and ploidy of the tumors remained unaltered. The results indicate that, although the tumor cells are stimulated by PDGF-AB in vitro, the in vivo milieu or tumor growth pattern may render the tumors less susceptible to exogenously administered PDGF-AB in vivo.

Collaboration


Dive into the Thomas M. Walz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zakaria Einbeigi

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge